1
|
Zhang C, Xu J, Wu Y, Xu C, Xu P. Base Editors-Mediated Gene Therapy in Hematopoietic Stem Cells for Hematologic Diseases. Stem Cell Rev Rep 2024; 20:1387-1405. [PMID: 38644403 PMCID: PMC11319617 DOI: 10.1007/s12015-024-10715-5] [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] [Accepted: 03/25/2024] [Indexed: 04/23/2024]
Abstract
Base editors, developed from the CRISPR/Cas system, consist of components such as deaminase and Cas variants. Since their emergence in 2016, the precision, efficiency, and safety of base editors have been gradually optimized. The feasibility of using base editors in gene therapy has been demonstrated in several disease models. Compared with the CRISPR/Cas system, base editors have shown great potential in hematopoietic stem cells (HSCs) and HSC-based gene therapy, because they do not generate double-stranded breaks (DSBs) while achieving the precise realization of single-base substitutions. This precise editing mechanism allows for the permanent correction of genetic defects directly at their source within HSCs, thus promising a lasting therapeutic effect. Recent advances in base editors are expected to significantly increase the number of clinical trials for HSC-based gene therapies. In this review, we summarize the development and recent progress of DNA base editors, discuss their applications in HSC gene therapy, and highlight the prospects and challenges of future clinical stem cell therapies.
Collapse
Affiliation(s)
- Chengpeng Zhang
- Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Hematology, Soochow Medical College, Soochow University, Suzhou, 215123, Jiangsu Province, China
| | - Jinchao Xu
- Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Hematology, Soochow Medical College, Soochow University, Suzhou, 215123, Jiangsu Province, China
| | - Yikang Wu
- Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Hematology, Soochow Medical College, Soochow University, Suzhou, 215123, Jiangsu Province, China
| | - Can Xu
- Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Hematology, Soochow Medical College, Soochow University, Suzhou, 215123, Jiangsu Province, China
| | - Peng Xu
- Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Hematology, Soochow Medical College, Soochow University, Suzhou, 215123, Jiangsu Province, China.
| |
Collapse
|
2
|
Saha A, Palchaudhuri R, Lanieri L, Hyzy S, Riddle MJ, Panthera J, Eide CR, Tolar J, Panoskaltsis-Mortari A, Gorfinkel L, Tkachev V, Gerdemann U, Alvarez-Calderon F, Palato ER, MacMillan ML, Wagner JE, Kean LS, Osborn MJ, Kiem HP, Scadden DT, Olson LM, Blazar BR. Alloengraftment without significant toxicity or GVHD in CD45 antibody-drug conjugate-conditioned Fanconi anemia mice. Blood 2024; 143:2201-2216. [PMID: 38447038 PMCID: PMC11143525 DOI: 10.1182/blood.2023023549] [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: 12/07/2023] [Revised: 02/09/2024] [Accepted: 02/25/2024] [Indexed: 03/08/2024] Open
Abstract
ABSTRACT Fanconi anemia (FA) is an inherited DNA repair disorder characterized by bone marrow (BM) failure, developmental abnormalities, myelodysplasia, leukemia, and solid tumor predisposition. Allogeneic hematopoietic stem cell transplantation (allo-HSCT), a mainstay treatment, is limited by conditioning regimen-related toxicity and graft-versus-host disease (GVHD). Antibody-drug conjugates (ADCs) targeting hematopoietic stem cells (HSCs) can open marrow niches permitting donor stem cell alloengraftment. Here, we report that single dose anti-mouse CD45-targeted ADC (CD45-ADC) facilitated stable, multilineage chimerism in 3 distinct FA mouse models representing 90% of FA complementation groups. CD45-ADC profoundly depleted host stem cell enriched Lineage-Sca1+cKit+ cells within 48 hours. Fanca-/- recipients of minor-mismatched BM and single dose CD45-ADC had peripheral blood (PB) mean donor chimerism >90%; donor HSCs alloengraftment was verified in secondary recipients. In Fancc-/- and Fancg-/- recipients of fully allogeneic grafts, PB mean donor chimerism was 60% to 80% and 70% to 80%, respectively. The mean percent donor chimerism in BM and spleen mirrored PB results. CD45-ADC-conditioned mice did not have clinical toxicity. A transient <2.5-fold increase in hepatocellular enzymes and mild-to-moderate histopathological changes were seen. Under GVHD allo-HSCT conditions, wild-type and Fanca-/- recipients of CD45-ADC had markedly reduced GVHD lethality compared with lethal irradiation. Moreover, single dose anti-human CD45-ADC given to rhesus macaque nonhuman primates on days -6 or -10 was at least as myeloablative as lethal irradiation. These data suggest that CD45-ADC can potently promote donor alloengraftment and hematopoiesis without significant toxicity or severe GVHD, as seen with lethal irradiation, providing strong support for clinical trial considerations in highly vulnerable patients with FA.
Collapse
Affiliation(s)
- Asim Saha
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics and Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | | | | | | | - Megan J. Riddle
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics and Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Jamie Panthera
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics and Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Cindy R. Eide
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics and Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Jakub Tolar
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics and Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Angela Panoskaltsis-Mortari
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics and Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Lev Gorfinkel
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, MA
| | - Victor Tkachev
- Massachusetts General Hospital Center for Transplantation Sciences, Mass General Brigham and Massachusetts General Hospital, Boston, MA
| | - Ulrike Gerdemann
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, MA
| | | | | | - Margaret L. MacMillan
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics and Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - John E. Wagner
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics and Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Leslie S. Kean
- Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, MA
| | - Mark J. Osborn
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics and Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Hans-Peter Kiem
- Department of Medicine, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA
| | - David T. Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA
| | | | - Bruce R. Blazar
- Division of Blood and Marrow Transplant & Cellular Therapy, Department of Pediatrics and Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| |
Collapse
|
3
|
Liu XL, Zhao J, Di XM, Cao G, Zhang H, Wang J. Case report: Highly response to low-dose brachytherapy in recurrent retroperitoneal leiomyosarcoma with FANCD2 frameshift mutation: a unique case study. Front Oncol 2024; 14:1339955. [PMID: 38634045 PMCID: PMC11021697 DOI: 10.3389/fonc.2024.1339955] [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: 11/20/2023] [Accepted: 03/12/2024] [Indexed: 04/19/2024] Open
Abstract
We report a case of recurrent retroperitoneal leiomyosarcoma in a male who achieved a rapid and robust but transient clinical response to low-dose iodine-125 brachytherapy. A FANCD2 frameshift mutation was detected by gene sequencing in the cancerous tissue.
Collapse
Affiliation(s)
| | | | | | | | - Hongtao Zhang
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | | |
Collapse
|
4
|
Borges MLRDR, Souza JLC, Rodrigues LH, Cornélio MTMN, Anjos ACD, Santos N, Salles TDJM. Clinical and cytogenetic profile of Fanconi anemia diagnosed after implementation of mitomycin C cytogenetic test in the state of Pernambuco, Brazil. Hematol Transfus Cell Ther 2024; 46:113-118. [PMID: 36759292 DOI: 10.1016/j.htct.2022.11.011] [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: 01/25/2022] [Revised: 08/22/2022] [Accepted: 11/16/2022] [Indexed: 01/30/2023] Open
Abstract
INTRODUCTION Fanconi anemia (FA) is a rare autosomal recessive disease characterized by chromosomal instability and increased predisposition to malignancy. The diagnosis of FA requires clinical evaluation, confirmation of chromosomal fragility and/or analysis of genetic mutations. Therefore, this study aims to identify the clinical profile of patients with FA in the state of Pernambuco, Brazil. METHOD We analyzed 100 individuals referred from the major hematology and bone marrow (BM) transplant centers in the state of Pernambuco, Brazil, between the years 2018 and 2022. The diagnosis of FA was performed using the mitomycin C chromosomal fragility test, clinical data and classical and molecular cytogenetic analyses. RESULTS We enrolled a total of 16 patients with FA to comprise this study. Most of these individuals (87.5%) came from the Agreste and Sertão regions of Pernambuco. We observed a slight female prevalence of FA (1.3:1). The primary clinical and laboratory findings were café au lait spots (62.5%) and bone abnormalities (53%, mainly thumb deformities [40%]). We performed BM cytogenetic analysis for eight patients - seven showed no chromosomal abnormalities and one presented the karyotype 47,XY,+21 [15]. CONCLUSIONS Our results are important to promote public health measures for the early diagnosis of FA, as well as to foster the engagement of a multidisciplinary group in the treatment of this disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Neide Santos
- Universidade Federal de Pernambuco (UFP), Recife, PE, Brazil
| | | |
Collapse
|
5
|
Mu A, Hira A, Mori M, Okamoto Y, Takata M. Fanconi anemia and Aldehyde Degradation Deficiency Syndrome: Metabolism and DNA repair protect the genome and hematopoiesis from endogenous DNA damage. DNA Repair (Amst) 2023; 130:103546. [PMID: 37572579 DOI: 10.1016/j.dnarep.2023.103546] [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: 05/12/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/14/2023]
Abstract
We have identified a set of Japanese children with hypoplastic anemia caused by combined defects in aldehyde degrading enzymes ADH5 and ALDH2. Their clinical characteristics overlap with a hereditary DNA repair disorder, Fanconi anemia. Our discovery of this disorder, termed Aldehyde Degradation Deficiency Syndrome (ADDS), reinforces the notion that endogenously generated aldehydes exert genotoxic effects; thus, the coupled actions of metabolism and DNA repair are required to maintain proper hematopoiesis and health.
Collapse
Affiliation(s)
- Anfeng Mu
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Multilayer Network Research Unit, Research Coordination Alliance, Kyoto University, Kyoto, Japan
| | - Asuka Hira
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Minako Mori
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yusuke Okamoto
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Minoru Takata
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan; Multilayer Network Research Unit, Research Coordination Alliance, Kyoto University, Kyoto, Japan.
| |
Collapse
|
6
|
Wang R, Zhang J, Cui X, Wang S, Chen T, Niu Y, Du X, Kong J, Wang L, Jiang Y. Multimolecular characteristics and role of BRCA1 interacting protein C-terminal helicase 1 (BRIP1) in human tumors: a pan-cancer analysis. World J Surg Oncol 2023; 21:91. [PMID: 36907870 PMCID: PMC10010046 DOI: 10.1186/s12957-022-02877-8] [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: 03/04/2022] [Accepted: 12/09/2022] [Indexed: 03/13/2023] Open
Abstract
BACKGROUND The aberrant expression of BRIP1 was associated with several cancers; however, the panoramic picture of BRIP1 in human tumors remains unclear. This study aims to explore the pan-cancerous picture of the expression of BRIP1 across 33 human cancers. METHODS Based on the data from TCGA and GTEx, a series of bioinformatic analyses were applied to systematically explore the genetic landscape and biologic function of BRIP1 in 33 human tumors. RESULTS We observed prognosis-related differential BRIP1 expressions between various carcinomas and the corresponding normal tissues. "Basal transcription factors," "homologous recombination," "nucleotide excision repair," and DNA metabolism pathways may play a role in the functional mechanisms of BRIP1. Patients with uterine corpus endometrial carcinoma presented with the highest alteration frequency of BRIP1 (nearly 10%). Single-nucleotide and copy number variations of BRIP1 were noticed in multiple cancers, and the expression of BRIP1 is significantly regulated by copy number variation in breast invasive carcinoma and lung squamous cell carcinoma. BRIP1 expression is negatively correlated with the DNA methylation levels in many tumors and is associated with the activation of apoptosis, cell cycle, DNA damage response, and inhibition of hormone ER and RNS/MARK signaling pathways. Moreover, a positive correlation was observed between BRIP1 expression and the immune infiltration levels of cancer-associated fibroblasts and CD8+ T cells in lung adenocarcinoma. CONCLUSION Our pan-cancer analysis of BRIP1 provides a valuable resource for understanding the multimolecular characteristics and biological function of BRIP1 across human cancers.
Collapse
Affiliation(s)
- Ruohuang Wang
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China
| | - Jisheng Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China
| | - Xin Cui
- Qingdao Women and Children's Hospital, Qingdao University, Qingdao, Shandong, 266000, China
| | - Shun Wang
- The Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200000, China
| | - Ting Chen
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China
| | - Yanfang Niu
- Department of Clinical Laboratory, Yuncheng Central Hospital, Yuncheng, Shanxi, 044000, China
| | - Xiaoyun Du
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China
| | - Jingwen Kong
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China
| | - Lin Wang
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China.
| | - Yan Jiang
- Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, 266000, China.
| |
Collapse
|
7
|
Visibelli A, Roncaglia B, Spiga O, Santucci A. The Impact of Artificial Intelligence in the Odyssey of Rare Diseases. Biomedicines 2023; 11:887. [PMID: 36979866 PMCID: PMC10045927 DOI: 10.3390/biomedicines11030887] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/28/2023] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
Emerging machine learning (ML) technologies have the potential to significantly improve the research and treatment of rare diseases, which constitute a vast set of diseases that affect a small proportion of the total population. Artificial Intelligence (AI) algorithms can help to quickly identify patterns and associations that would be difficult or impossible for human analysts to detect. Predictive modeling techniques, such as deep learning, have been used to forecast the progression of rare diseases, enabling the development of more targeted treatments. Moreover, AI has also shown promise in the field of drug development for rare diseases with the identification of subpopulations of patients who may be most likely to respond to a particular drug. This review aims to highlight the achievements of AI algorithms in the study of rare diseases in the past decade and advise researchers on which methods have proven to be most effective. The review will focus on specific rare diseases, as defined by a prevalence rate that does not exceed 1-9/100,000 on Orphanet, and will examine which AI methods have been most successful in their study. We believe this review can guide clinicians and researchers in the successful application of ML in rare diseases.
Collapse
Affiliation(s)
- Anna Visibelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Bianca Roncaglia
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Ottavia Spiga
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- Competence Center ARTES 4.0, 53100 Siena, Italy
- SienabioACTIVE—SbA, 53100 Siena, Italy
| | - Annalisa Santucci
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- Competence Center ARTES 4.0, 53100 Siena, Italy
- SienabioACTIVE—SbA, 53100 Siena, Italy
| |
Collapse
|
8
|
Pulicari F, Pellegrini M, Scribante A, Kuhn E, Spadari F. Pathological Background and Clinical Procedures in Oral Surgery Haemostasis Disorders: A Narrative Review. APPLIED SCIENCES 2023; 13:2076. [DOI: 10.3390/app13042076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Haemostasis disorders are serious pathologies that could put dental and surgical procedures at risk as they are associated with postoperative bleeding, which in some circumstances could be prolonged and dangerous for the patient. In-depth knowledge of the problems associated with coagulation pathologies and the suitable specific procedures should be implemented in dental practice. A good awareness of the clinical protocols to be used in these circumstances may help reduce operator stress and increase patient compliance. Collaboration with the haematologist is always recommended to establish an adequate treatment plan, both regarding the administration of therapies that promote haemostasis and for assessing the operative risk. Hereby, we summarize the congenital and hereditary pathologies that lead to haemostasis disorders, which can be found in patients undergoing dental procedures. The purpose of this narrative review is to frame the diseases from a clinical, anamnestic, and etiopathological standpoint, as well as to evaluate an operative approach to the pathology under consideration, with particular attention to anaesthesia manoeuvres and post-surgical haemostasis, to avoid hematoma formation and uncontrolled bleeding which can lead procedure failure up and even death. Of note, it is likewise important to educate the patient about prevention, to keep the oral cavity healthy and avoid invasive procedures, limiting the number of operative sessions.
Collapse
Affiliation(s)
- Federica Pulicari
- Department of Biomedical Surgical and Dental Sciences, University of Milan, Via Della Commenda 10, 20122 Milan, Italy
- Maxillo-Facial and Odontostomatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Matteo Pellegrini
- Department of Biomedical Surgical and Dental Sciences, University of Milan, Via Della Commenda 10, 20122 Milan, Italy
- Maxillo-Facial and Odontostomatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Section of Dentistry, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Andrea Scribante
- Section of Dentistry, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| | - Elisabetta Kuhn
- Department of Biomedical Surgical and Dental Sciences, University of Milan, Via Della Commenda 10, 20122 Milan, Italy
- Pathology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesco Spadari
- Department of Biomedical Surgical and Dental Sciences, University of Milan, Via Della Commenda 10, 20122 Milan, Italy
- Maxillo-Facial and Odontostomatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
9
|
He R, Fan Q, Li Y, Zhu Q, Hu D, Du J, Xing Y, Li H, Liang X, Yang Y. Identification of Common and Specific Genes Involved in Mouse Models of Age-Related and Cyclophosphamide-Induced Diminished Ovarian Reserve. Reprod Sci 2022; 30:1965-1978. [PMID: 36587055 DOI: 10.1007/s43032-022-01161-0] [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: 07/26/2022] [Accepted: 12/22/2022] [Indexed: 01/02/2023]
Abstract
Diminished ovarian reserve (DOR) is an etiologically heterogeneous disorder that usually leads to poor reproductive outcomes. Does a specific or common pathogenesis exist for DOR subtypes with different etiologies? Two frequently used mouse models, age-related DOR (AR-DOR) and cyclophosphamide (CTX)-induced DOR (CTX-DOR), were successfully established, and RNA sequencing was performed on ovarian tissue samples. Differentially expressed genes (DEGs) in each subtype and common DEGs (co-DEGs) in the two subtypes were identified. Subsequently, we performed comprehensive bioinformatics analyses, including an evaluation of immune cell infiltration. Finally, the genes of interest were further validated by performing RT-qPCR and immunohistochemistry. In AR-DOR mice, functional enrichment analyses showed that upregulated DEGs were mainly involved in the inflammatory/immune response, while downregulated DEGs were involved in DNA damage repair. In CTX-DOR mice, the inflammatory/immune response and cell apoptosis played significant roles. Meanwhile, 406 co-DEGs were identified from the two models. The biological functions of these co-DEGs were associated with inflammatory/immune responses. The analysis of immune cell infiltration showed reduced infiltration of Treg cells, as well as increased infiltration of M0 macrophages, NK resting, and T cells CD4 follicular in both DOR subtypes. The results of the validation experiments were consistent with the RNA sequencing data. In conclusion, the inflammatory/immune response might be the common pathogenesis for the two DOR subtypes, while DNA repair and cell apoptosis may have different roles in the two subtypes. These results may provide potential insights for mechanistic research and therapeutic targets of DOR.
Collapse
Affiliation(s)
- Ruifen He
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Qigang Fan
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yi Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Qinying Zhu
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Dan Hu
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Junhong Du
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yijuan Xing
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Hongli Li
- Department of Obstetrics and Gynecology, Key Laboratory for Gynecologic Oncology Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, Key Laboratory for Gynecologic Oncology Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China.
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, Key Laboratory for Gynecologic Oncology Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
10
|
Jacobs K, Doerdelmann C, Krietsch J, González-Acosta D, Mathis N, Kushinsky S, Guarino E, Gómez-Escolar C, Martinez D, Schmid JA, Leary PJ, Freire R, Ramiro AR, Eischen CM, Mendez J, Lopes M. Stress-triggered hematopoietic stem cell proliferation relies on PrimPol-mediated repriming. Mol Cell 2022; 82:4176-4188.e8. [PMID: 36152632 PMCID: PMC10251193 DOI: 10.1016/j.molcel.2022.09.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 07/01/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022]
Abstract
Stem cell division is linked to tumorigenesis by yet-elusive mechanisms. The hematopoietic system reacts to stress by triggering hematopoietic stem and progenitor cell (HSPC) proliferation, which can be accompanied by chromosomal breakage in activated hematopoietic stem cells (HSCs). However, whether these lesions persist in their downstream progeny and induce a canonical DNA damage response (DDR) remains unclear. Inducing HSPC proliferation by simulated viral infection, we report that the associated DNA damage is restricted to HSCs and that proliferating HSCs rewire their DDR upon endogenous and clastogen-induced damage. Combining transcriptomics, single-cell and single-molecule assays on murine bone marrow cells, we found accelerated fork progression in stimulated HSPCs, reflecting engagement of PrimPol-dependent repriming, at the expense of replication fork reversal. Ultimately, competitive bone marrow transplantation revealed the requirement of PrimPol for efficient HSC amplification and bone marrow reconstitution. Hence, fine-tuning replication fork plasticity is essential to support stem cell functionality upon proliferation stimuli.
Collapse
Affiliation(s)
- Kurt Jacobs
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Cyril Doerdelmann
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Jana Krietsch
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Daniel González-Acosta
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; DNA Replication Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Nicolas Mathis
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Saul Kushinsky
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Estrella Guarino
- DNA Replication Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Carmen Gómez-Escolar
- B Lymphocyte Biology Laboratory, Spanish National Center for Cardiovascular Research (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Dolores Martinez
- Flow Cytometry Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Jonas A Schmid
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Peter J Leary
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Functional Genomic Center Zurich, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias, Tenerife, Spain; Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, Spain; Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Almudena R Ramiro
- B Lymphocyte Biology Laboratory, Spanish National Center for Cardiovascular Research (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Christine M Eischen
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Juan Mendez
- DNA Replication Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, 28029 Madrid, Spain.
| | - Massimo Lopes
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
11
|
Liu P, Lin C, Liu L, Lu Z, Tu Z, Liu H. RAD54B mutations enhance the sensitivity of ovarian cancer cells to poly(ADP-ribose) polymerase (PARP) inhibitors. J Biol Chem 2022; 298:102354. [PMID: 35952757 PMCID: PMC9463535 DOI: 10.1016/j.jbc.2022.102354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022] Open
Abstract
Synthetic lethal targeting of homologous recombination (HR)–deficient ovarian cancers (OvCas) with poly(ADP-ribose) polymerase inhibitors (PARPis) has attracted considerable attention. Olaparib was the first PARPi approved by the Food and Drug Administration, offering significant clinical benefits in BRCA1/2-deficient OvCas. However, only approximately 20% of OvCa patients harbor BRCA1/2 mutations. Given the shared roles that BRCA1/2 have with other HR regulators, alterations in HR genes may also contribute to “BRCAness profiles” in OvCas. RAD54B has been considered a key player in HR repair, although its roles and therapeutic potential in cancers need further investigation. Here, we identified 22 frequently mutated HR genes by whole-exome sequencing of OvCa tissues from 82 patients. To our surprise, 7.3% of patients were found to harbor mutations of RAD54B, the third-highest mutated gene among patients. We determined that RAD54B-mutated tumor tissues harbored more DNA double-strand breaks than normal tissues. Additionally, we found that RAD54B knockdown inhibited HR repair, enhanced sensitivities of OvCa cells with increased DNA double-strand breaks to olaparib, and induced apoptosis. Enhanced inhibitory effects of olaparib on the growth of ES2 xenograft tumors were further demonstrated by RAD54B knockdown. Finally, we show that restoration with wildtype RAD54B rather than RAD54BN593S and RAD54BH219Y, identified in patients, abolished the effects of RAD54B knockdown, indicating these RAD54B mutants probably malfunctioned in HR repair. Our investigations may offer insight into the contributions of RAD54B mutations to synthetic lethality with olaparib treatment in OvCas, enrich the gene list for “HR deficiency scoring,” and expand the applications of PARPis.
Collapse
Affiliation(s)
- Peng Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Chunxiu Lin
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Lanlan Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Ziwen Lu
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Zhigang Tu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | - Hanqing Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
12
|
Jang SW, Kim JM. The RPA inhibitor HAMNO sensitizes Fanconi anemia pathway-deficient cells. Cell Cycle 2022; 21:1468-1478. [PMID: 35506981 PMCID: PMC9278452 DOI: 10.1080/15384101.2022.2074200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The Fanconi anemia (FA) DNA repair pathway is required for DNA inter-strand crosslink (ICL) repair. Besides its role in ICL repair, FA proteins play a central role in stabilizing stalled replication forks, thereby ensuring genome integrity. We previously demonstrated that depletion of replication protein A (RPA) induces the activation of FA pathway leading to FANCD2 monoubiquitination and FANCD2 foci formation. Thus, we speculated that FA-deficient cells would be more sensitive to RPA inhibition compared to FA-proficient cells. Following treatment with RPA inhibitor HAMNO, we observed significant induction in FANCD2 monoubiquitination and foci formation as observed in RPA depletion. In addition, HAMNO treatment caused increased levels of ϒ-H2AX and S-phase accumulation in FA-deficient cells. Importantly, FA-deficient cells showed more increased sensitivity to HAMNO than FA-proficient cells. Moreover, in combination with cisplatin, HAMNO further enhanced the cytotoxicity of cisplatin in FA-deficient cells, while being less toxic against FA-proficient cells. This result suggests that RPA inhibition might be a potential therapeutic candidate for the treatment of FA pathway-deficient tumors.
Collapse
Affiliation(s)
- Seok-Won Jang
- Department of Pharmacology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
| | - Jung Min Kim
- Department of Pharmacology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
| |
Collapse
|
13
|
Ntemou E, Vidal PD, Alexandri C, Van den Steen G, Lambertini M, Demeestere I. Ovarian toxicity of carboplatin and paclitaxel in mouse carriers of mutation in BRIP1 tumor suppressor gene. Sci Rep 2022; 12:1658. [PMID: 35105904 PMCID: PMC8807594 DOI: 10.1038/s41598-022-05357-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/11/2022] [Indexed: 11/19/2022] Open
Abstract
More than 10% of women diagnosed with breast cancer during reproductive age carry hereditary germline pathogenic variants in high-penetrance BRCA genes or in others genes involved in DNA repair mechanisms such as PALB2, BRIP or ATM. Anticancer treatments may have an additional negative impact on the ovarian reserve and subsequently on the fertility of young patients carrying such mutations. Recently, the combination of carboplatin and paclitaxel is being recommended to these BRCA-mutated patients as neoadjuvant therapy. However, the impact on the ovary is unknown. Here, we investigated their effect of on the ovarian reserve using mice carriers of BRCA1-interacting protein C-terminal helicase-1 (BRIP1) mutation that plays an important role in BRCA1-dependent DNA repair. Results revealed that the administration of carboplatin or paclitaxel did not affect the ovarian reserve although increased DNA double-strand breaks were observed with carboplatin alone. Co-administration of carboplatin and paclitaxel resulted in a significant reduction of the ovarian reserve leading to a lower IVF performance, and an activation of the PI3K-Pten pathway, irrespective of the genetic background. This study suggests that co-administration of carboplatin and paclitaxel induces cumulative ovarian damage and infertility but a heterozygote genetic predisposition for DNA damage related to BRCA1 gene function does not increase this risk.
Collapse
Affiliation(s)
- E Ntemou
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - P Diaz Vidal
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - C Alexandri
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - G Van den Steen
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - M Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy
- Department of Medical Oncology, UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - I Demeestere
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium.
- Obstetrics and Gynaecology Department, Erasme Hospital, Brussels, Belgium.
| |
Collapse
|
14
|
Maia N, Nabais Sá MJ, Oliveira C, Santos F, Soares CA, Prior C, Tkachenko N, Santos R, de Brouwer APM, Jacome A, Porto B, Jorge P. Can the Synergic Contribution of Multigenic Variants Explain the Clinical and Cellular Phenotypes of a Neurodevelopmental Disorder? Genes (Basel) 2021; 13:78. [PMID: 35052418 PMCID: PMC8774836 DOI: 10.3390/genes13010078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/14/2021] [Accepted: 12/23/2021] [Indexed: 11/16/2022] Open
Abstract
We describe an infant female with a syndromic neurodevelopmental clinical phenotype and increased chromosome instability as cellular phenotype. Genotype characterization revealed heterozygous variants in genes directly or indirectly linked to DNA repair: a de novo X-linked HDAC8 pathogenic variant, a paternally inherited FANCG pathogenic variant and a maternally inherited BRCA2 variant of uncertain significance. The full spectrum of the phenotype cannot be explained by any of the heterozygous variants on their own; thus, a synergic contribution is proposed. Complementation studies showed that the FANCG gene from the Fanconi Anaemia/BRCA (FA/BRCA) DNA repair pathway was impaired, indicating that the variant in FANCG contributes to the cellular phenotype. The patient's chromosome instability represents the first report where heterozygous variant(s) in the FA/BRCA pathway are implicated in the cellular phenotype. We propose that a multigenic contribution of heterozygous variants in HDAC8 and the FA/BRCA pathway might have a role in the phenotype of this neurodevelopmental disorder. The importance of these findings may have repercussion in the clinical management of other cases with a similar synergic contribution of heterozygous variants, allowing the establishment of new genotype-phenotype correlations and motivating the biochemical study of the underlying mechanisms.
Collapse
Affiliation(s)
- Nuno Maia
- Unidade de Genética Molecular, Centro de Genética Médica Jacinto de Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal; (N.M.); (F.S.); (R.S.)
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Laboratory for Integrative and Translational Research in Population Health (ITR), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal; (M.J.N.S.); (C.A.S.)
| | - Maria João Nabais Sá
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Laboratory for Integrative and Translational Research in Population Health (ITR), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal; (M.J.N.S.); (C.A.S.)
| | - Cláudia Oliveira
- Laboratório Citogenética, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal; (C.O.); (B.P.)
| | - Flávia Santos
- Unidade de Genética Molecular, Centro de Genética Médica Jacinto de Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal; (N.M.); (F.S.); (R.S.)
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Laboratory for Integrative and Translational Research in Population Health (ITR), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal; (M.J.N.S.); (C.A.S.)
| | - Célia Azevedo Soares
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Laboratory for Integrative and Translational Research in Population Health (ITR), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal; (M.J.N.S.); (C.A.S.)
- Serviço de Genética Médica, Centro de Genética Médica Doutor Jacinto Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal;
| | - Catarina Prior
- Unidade de Neurodesenvolvimento do Serviço de Pediatria do Centro Materno-Infantil do Norte (CMIN), Centro Hospitalar Universitário do Porto (CHUPorto), 4050-651 Porto, Portugal;
| | - Nataliya Tkachenko
- Serviço de Genética Médica, Centro de Genética Médica Doutor Jacinto Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal;
| | - Rosário Santos
- Unidade de Genética Molecular, Centro de Genética Médica Jacinto de Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal; (N.M.); (F.S.); (R.S.)
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Laboratory for Integrative and Translational Research in Population Health (ITR), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal; (M.J.N.S.); (C.A.S.)
| | - Arjan P. M. de Brouwer
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, 6525 GA Nijmegen, The Netherlands;
| | - Ariana Jacome
- Chromosome Instability and Dynamics Lab. (CID), Instituto de Inovação e Investigação (i3S), Universidade do Porto, 4200-135 Porto, Portugal;
| | - Beatriz Porto
- Laboratório Citogenética, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal; (C.O.); (B.P.)
| | - Paula Jorge
- Unidade de Genética Molecular, Centro de Genética Médica Jacinto de Magalhães (CGM), Centro Hospitalar Universitário do Porto (CHUPorto), 4099-028 Porto, Portugal; (N.M.); (F.S.); (R.S.)
- Unit for Multidisciplinary Research in Biomedicine (UMIB), Laboratory for Integrative and Translational Research in Population Health (ITR), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal; (M.J.N.S.); (C.A.S.)
| |
Collapse
|
15
|
Kontandreopoulou CN, Diamantopoulos PT, Tiblalexi D, Giannakopoulou N, Viniou NA. PARP1 as a therapeutic target in acute myeloid leukemia and myelodysplastic syndrome. Blood Adv 2021; 5:4794-4805. [PMID: 34529761 PMCID: PMC8759124 DOI: 10.1182/bloodadvances.2021004638] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/15/2021] [Indexed: 12/31/2022] Open
Abstract
Poly(ADP-ribose) polymerase 1 (PARP1) is a key mediator of various forms of DNA damage repair and plays an important role in the progression of several cancer types. The enzyme is activated by binding to DNA single-strand and double-strand breaks. Its contribution to chromatin remodeling makes PARP1 crucial for gene expression regulation. Inhibition of its activity with small molecules leads to the synthetic lethal effect by impeding DNA repair in the treatment of cancer cells. At first, PARP1 inhibitors (PARPis) were developed to target breast cancer mutated cancer cells. Currently, PARPis are being studied to be used in a broader variety of patients either as single agents or in combination with chemotherapy, antiangiogenic agents, ionizing radiation, and immune checkpoint inhibitors. Ongoing clinical trials on olaparib, rucaparib, niraparib, veliparib, and the recent talazoparib show the advantage of these agents in overcoming PARPi resistance and underline their efficacy in targeted treatment of several hematologic malignancies. In this review, focusing on the crucial role of PARP1 in physiological and pathological effects in myelodysplastic syndrome and acute myeloid leukemia, we give an outline of the enzyme's mechanisms of action and its role in the pathophysiology and prognosis of myelodysplastic syndrome/acute myeloid leukemia and we analyze the available data on the use of PARPis, highlighting their promising advances in clinical application.
Collapse
Affiliation(s)
- Christina-Nefeli Kontandreopoulou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis T. Diamantopoulos
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina Tiblalexi
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nefeli Giannakopoulou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Nora-Athina Viniou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
16
|
Chang L, Gao X, Wang Y, Huang C, Gao M, Wang X, Liu C, Wu W, An W, Wan Y, Zhang A, Zhang Y, Yuan W, Zhu X. DNAH2 facilitates the homologous recombination repair of Fanconi anemia pathway through modulating FANCD2 ubiquitination. BLOOD SCIENCE 2021; 3:71-77. [PMID: 35402838 PMCID: PMC8974987 DOI: 10.1097/bs9.0000000000000076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 05/14/2021] [Indexed: 11/16/2022] Open
Abstract
Fanconi anemia (FA), an X-linked genetic or autosomal recessive disease, exhibits complicated pathogenesis. Previously, we detected the mutated Dynein Axonemal Heavy Chain 2 (DNAH2) gene in 2 FA cases. Herein, we further investigated the potential association between DNAH2 and the homologous recombination repair pathway of FA. The assays of homologous recombination repair, mitomycin C (MMC) sensitivity, immunofluorescence, and ubiquitination modification were performed in U2OS and DR-U2OS cell lines. In MMC-treated U2OS cells, the downregulation of the DNAH2 gene increased the sensitivity of cells to DNA inter-strand crosslinks. We also observed the reduced enrichment of FANCD2 protein to DNA damage sites. Furthermore, the ubiquitination modification level of FANCD2 was influenced by the deficiency of DNAH2. Thus, our results suggest that DNAH2 may modulate the cell homologous recombination repair partially by increasing the ubiquitination and the enrichment to DNA damage sites of FANCD2. DNAH2 may act as a novel co-pathogenic gene of FA patients.
Collapse
Affiliation(s)
- Lixian Chang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xingjie Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuxia Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Department of Hematology, Myelodysplastic Syndromes Diagnosis and Therapy Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunmin Huang
- Beijing Institute of Genomics, Chinese Academy of Sciences (China National Center for Bioinformation), Beijing, China
| | - Min Gao
- Beijing Institute of Genomics, Chinese Academy of Sciences (China National Center for Bioinformation), Beijing, China
| | - Xiaomin Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Chao Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wenqi Wu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wenbin An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yang Wan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Aoli Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yingchi Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Weiping Yuan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
17
|
Alternative DNA Structures In Vivo: Molecular Evidence and Remaining Questions. Microbiol Mol Biol Rev 2020; 85:85/1/e00110-20. [PMID: 33361270 DOI: 10.1128/mmbr.00110-20] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Duplex DNA naturally folds into a right-handed double helix in physiological conditions. Some sequences of unusual base composition may nevertheless form alternative structures, as was shown for many repeated sequences in vitro However, evidence for the formation of noncanonical structures in living cells is difficult to gather. It mainly relies on genetic assays demonstrating their function in vivo or through genetic instability reflecting particular properties of such structures. Efforts were made to reveal their existence directly in a living cell, mainly by generating antibodies specific to secondary structures or using chemical ligands selected for their affinity to these structures. Among secondary structure-forming DNAs are G-quadruplexes, human fragile sites containing minisatellites, AT-rich regions, inverted repeats able to form cruciform structures, hairpin-forming CAG/CTG triplet repeats, and triple helices formed by homopurine-homopyrimidine GAA/TTC trinucleotide repeats. Many of these alternative structures are involved in human pathologies, such as neurological or developmental disorders, as in the case of trinucleotide repeats, or cancers triggered by translocations linked to fragile sites. This review will discuss and highlight evidence supporting the formation of alternative DNA structures in vivo and will emphasize the role of the mismatch repair machinery in binding mispaired DNA duplexes, triggering genetic instability.
Collapse
|
18
|
Turan V, Oktay K. BRCA-related ATM-mediated DNA double-strand break repair and ovarian aging. Hum Reprod Update 2020; 26:43-57. [PMID: 31822904 DOI: 10.1093/humupd/dmz043] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/26/2019] [Accepted: 11/05/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Oocyte aging has significant clinical consequences, and yet no treatment exists to address the age-related decline in oocyte quality. The lack of progress in the treatment of oocyte aging is due to the fact that the underlying molecular mechanisms are not sufficiently understood. BRCA1 and 2 are involved in homologous DNA recombination and play essential roles in ataxia telangiectasia mutated (ATM)-mediated DNA double-strand break (DSB) repair. A growing body of laboratory, translational and clinical evidence has emerged within the past decade indicating a role for BRCA function and ATM-mediated DNA DSB repair in ovarian aging. OBJECTIVE AND RATIONALE Although there are several competing or complementary theories, given the growing evidence tying BRCA function and ATM-mediated DNA DSB repair mechanisms in general to ovarian aging, we performed this review encompassing basic, translational and clinical work to assess the current state of knowledge on the topic. A clear understanding of the mechanisms underlying oocyte aging may result in targeted treatments to preserve ovarian reserve and improve oocyte quality. SEARCH METHODS We searched for published articles in the PubMed database containing key words, BRCA, BRCA1, BRCA2, Mutations, Fertility, Ovarian Reserve, Infertility, Mechanisms of Ovarian Aging, Oocyte or Oocyte DNA Repair, in the English-language literature until May 2019. We did not include abstracts or conference proceedings, with the exception of our own. OUTCOMES Laboratory studies provided robust and reproducible evidence that BRCA1 function and ATM-mediated DNA DSB repair, in general, weakens with age in oocytes of multiple species including human. In both women with BRCA mutations and BRCA-mutant mice, primordial follicle numbers are reduced and there is accelerated accumulation of DNA DSBs in oocytes. In general, women with BRCA1 mutations have lower ovarian reserves and experience earlier menopause. Laboratory evidence also supports critical role for BRCA1 and other ATM-mediated DNA DSB repair pathway members in meiotic function. When laboratory, translational and clinical evidence is considered together, BRCA-related ATM-mediated DNA DSB repair function emerges as a likely regulator of ovarian aging. Moreover, DNA damage and repair appear to be key features in chemotherapy-induced ovarian aging. WIDER IMPLICATIONS The existing data suggest that the BRCA-related ATM-mediated DNA repair pathway is a strong candidate to be a regulator of oocyte aging, and the age-related decline of this pathway likely impairs oocyte health. This knowledge may create an opportunity to develop targeted treatments to reverse or prevent physiological or chemotherapy-induced oocyte aging. On the immediate practical side, women with BRCA or similar mutations may need to be specially counselled for fertility preservation.
Collapse
Affiliation(s)
- Volkan Turan
- Department of Obstetrics and Gynecology, Uskudar University School of Medicine, Istanbul, Turkey.,Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Kutluk Oktay
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
19
|
Dillon B, Feben C, Segal D, du Plessis J, Reynders D, Wainwright R, Poole J, Krause A. Endocrine profiling in patients with Fanconi anemia, homozygous for a FANCG founder mutation. Mol Genet Genomic Med 2020; 8:e1351. [PMID: 32529760 PMCID: PMC7434606 DOI: 10.1002/mgg3.1351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Fanconi anemia (FA) is phenotypically diverse, hereditary condition associated with bone marrow failure, multiple physical abnormalities, and an increased susceptibility to the development of malignancies. Less recognized manifestations of FA include endocrine abnormalities. International discourse has highlighted that these abnormalities are widespread among children and adults with FA. To date there has been no systematic study that has evaluated the endocrine abnormalities in a cohort of patients with FA, homozygous for a founder mutation (c.637_643del (p.Tyr213Lysfs*6)) in FANCG. The objectives of the study were to evaluate endocrine gland function in patients with FA of a single FA genotype, and to determine the frequency and nature of endocrine abnormalities in this group. METHODS Cross-sectional, descriptive study of 24 South African patients of African ancestry with FA (homozygous for a FANCG founder mutation). Outcomes measured included growth, pubertal status, growth hormone axis screening, thyroid gland function, glucose and insulin metabolism and bone age (BA). RESULTS Endocrine dysfunction was present in 70.8% (17 of 24), including abnormal insulin-like growth factor 1 (IGF-1)/insulin-like growth factor-binding protein 3 (IGFBP-3) in 25.0% (6 of 24), insulin resistance in 41.7% (10 of 24), abnormal thyroid function in 16.7% (4 of 24) and short stature in 45.8% (11 of 24). No abnormalities of glucose metabolism were identified. Abnormal pubertal status was seen in three males (12.5%). Abnormal BAs were present in 34.8% (8 of 23). CONCLUSION Endocrine abnormalities occur at a high frequency in patients with FA, homozygous for a FANCG founder mutation, similar to other FA cohorts. Our data are specific to FA patients with a single genotype, and therefore provide the first genotype-phenotype information on endocrine abnormalities in South African patients, homozygous for a FANCG founder mutation. Recommendations regarding endocrine screening in this patient subgroup are made, including, but not limited to, baseline testing of thyroid function, fasted insulin and glucose, and IGF-1 and IGFBP-3.
Collapse
Affiliation(s)
- Bronwyn Dillon
- Division of Human GeneticsNational Health Laboratory Service and School of PathologyFaculty of Health SciencesThe University of the WitwatersrandJohannesburgGautengSouth Africa
| | - Candice Feben
- Division of Human GeneticsNational Health Laboratory Service and School of PathologyFaculty of Health SciencesThe University of the WitwatersrandJohannesburgGautengSouth Africa
| | - David Segal
- The University of the Witwatersrand and The Wits University Donald Gordon Medical CentreJohannesburgGautengSouth Africa
| | - Johannes du Plessis
- Paediatric Oncology UnitUniversitas Hospital and The University of the Free StateBloemfonteinFree StateSouth Africa
| | - David Reynders
- Paediatric Haematology and Oncology UnitSteve Biko Academic Hospital and The University of PretoriaPretoriaGautengSouth Africa
| | - Rosalind Wainwright
- Department of Paediatric Haematology and OncologyChris Hani Baragwanath Academic Hospital and The University of the WitwatersrandJohannesburgGautengSouth Africa
| | - Janet Poole
- Department of Paediatric Haematology and OncologyCharlotte Maxeke Johannesburg Academic Hospital and The University of the WitwatersrandJohannesburgGautengSouth Africa
| | - Amanda Krause
- Division of Human GeneticsNational Health Laboratory Service and School of PathologyFaculty of Health SciencesThe University of the WitwatersrandJohannesburgGautengSouth Africa
| |
Collapse
|
20
|
Rageul J, Kim H. Fanconi anemia and the underlying causes of genomic instability. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:693-708. [PMID: 31983075 PMCID: PMC7778457 DOI: 10.1002/em.22358] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/03/2020] [Accepted: 01/21/2020] [Indexed: 05/02/2023]
Abstract
Fanconi anemia (FA) is a rare genetic disorder, characterized by birth defects, progressive bone marrow failure, and a predisposition to cancer. This devastating disease is caused by germline mutations in any one of the 22 known FA genes, where the gene products are primarily responsible for the resolution of DNA interstrand cross-links (ICLs), a type of DNA damage generally formed by cytotoxic chemotherapeutic agents. However, the identity of endogenous mutagens that generate DNA ICLs remains largely elusive. In addition, whether DNA ICLs are indeed the primary cause behind FA phenotypes is still a matter of debate. Recent genetic studies suggest that naturally occurring reactive aldehydes are a primary source of DNA damage in hematopoietic stem cells, implicating that they could play a role in genome instability and FA. Emerging lines of evidence indicate that the FA pathway constitutes a general surveillance mechanism for the genome by protecting against a variety of DNA replication stresses. Therefore, understanding the DNA repair signaling that is regulated by the FA pathway, and the types of DNA lesions underlying the FA pathophysiology is crucial for the treatment of FA and FA-associated cancers. Here, we review recent advances in our understanding of the relationship between reactive aldehydes, bone marrow dysfunction, and FA biology in the context of signaling pathways triggered during FA-mediated DNA repair and maintenance of the genomic integrity. Environ. Mol. Mutagen. 2020. © 2020 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Julie Rageul
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794, USA
| | - Hyungjin Kim
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794, USA
- Stony Brook Cancer Center, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York 11794, USA
- Correspondence to: Hyungjin Kim, Ph.D., Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Basic Sciences Tower 8-125, 100 Nicolls Rd., Stony Brook, NY 11794, Phone: 631-444-3134, FAX: 631-444-3218,
| |
Collapse
|
21
|
The Role of Ataxia Telangiectasia Mutant and Rad3-Related DNA Damage Response in Pathogenesis of Human Papillomavirus. Pathogens 2020; 9:pathogens9060506. [PMID: 32585979 PMCID: PMC7350315 DOI: 10.3390/pathogens9060506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 12/16/2022] Open
Abstract
Human papillomavirus (HPV) infection leads to a variety of benign lesions and malignant tumors such as cervical cancer and head and neck squamous cell carcinoma. Several HPV vaccines have been developed that can help to prevent cervical carcinoma, but these vaccines are only effective in individuals with no prior HPV infection. Thus, it is still important to understand the HPV life cycle and in particular the association of HPV with human pathogenesis. HPV production requires activation of the DNA damage response (DDR), which is a complex signaling network composed of multiple sensors, mediators, transducers, and effectors that safeguard cellular DNAs to maintain the host genome integrity. In this review, we focus on the roles of the ataxia telangiectasia mutant and Rad3-related (ATR) DNA damage response in HPV DNA replication. HPV can induce ATR expression and activate the ATR pathway. Inhibition of the ATR pathway results in suppression of HPV genome maintenance and amplification. The mechanisms underlying this could be through various molecular pathways such as checkpoint signaling and transcriptional regulation. In light of these findings, other downstream mechanisms of the ATR pathway need to be further investigated for better understanding HPV pathogenesis and developing novel ATR DDR-related inhibitors against HPV infection.
Collapse
|
22
|
Kreutmair S, Erlacher M, Andrieux G, Istvanffy R, Mueller-Rudorf A, Zwick M, Rückert T, Pantic M, Poggio T, Shoumariyeh K, Mueller TA, Kawaguchi H, Follo M, Klingeberg C, Wlodarski M, Baumann I, Pfeifer D, Kulinski M, Rudelius M, Lemeer S, Kuster B, Dierks C, Peschel C, Cabezas-Wallscheid N, Duque-Afonso J, Zeiser R, Cleary ML, Schindler D, Schmitt-Graeff A, Boerries M, Niemeyer CM, Oostendorp RA, Duyster J, Illert AL. Loss of the Fanconi anemia-associated protein NIPA causes bone marrow failure. J Clin Invest 2020; 130:2827-2844. [PMID: 32338640 PMCID: PMC7260023 DOI: 10.1172/jci126215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Inherited bone marrow failure syndromes (IBMFSs) are a heterogeneous group of disorders characterized by defective hematopoiesis, impaired stem cell function, and cancer susceptibility. Diagnosis of IBMFS presents a major challenge due to the large variety of associated phenotypes, and novel, clinically relevant biomarkers are urgently needed. Our study identified nuclear interaction partner of ALK (NIPA) as an IBMFS gene, as it is significantly downregulated in a distinct subset of myelodysplastic syndrome-type (MDS-type) refractory cytopenia in children. Mechanistically, we showed that NIPA is major player in the Fanconi anemia (FA) pathway, which binds FANCD2 and regulates its nuclear abundance, making it essential for a functional DNA repair/FA/BRCA pathway. In a knockout mouse model, Nipa deficiency led to major cell-intrinsic defects, including a premature aging phenotype, with accumulation of DNA damage in hematopoietic stem cells (HSCs). Induction of replication stress triggered a reduction in and functional decline of murine HSCs, resulting in complete bone marrow failure and death of the knockout mice with 100% penetrance. Taken together, the results of our study add NIPA to the short list of FA-associated proteins, thereby highlighting its potential as a diagnostic marker and/or possible target in diseases characterized by hematopoietic failure.
Collapse
Affiliation(s)
- Stefanie Kreutmair
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Miriam Erlacher
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, and
| | - Geoffroy Andrieux
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, University Medical Center — University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Rouzanna Istvanffy
- Department of Internal Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Alina Mueller-Rudorf
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melissa Zwick
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tamina Rückert
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Milena Pantic
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Teresa Poggio
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Khalid Shoumariyeh
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tony A. Mueller
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hiroyuki Kawaguchi
- Department of Pediatrics, National Defense Medical College, Saitama, Japan
| | - Marie Follo
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cathrin Klingeberg
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marcin Wlodarski
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, and
| | - Irith Baumann
- Institute of Pathology, Health Center Böblingen, Böblingen, Germany
| | - Dietmar Pfeifer
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michal Kulinski
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Martina Rudelius
- Institute of Pathology, Ludwig Maximilian University Munich, Munich, Germany
| | - Simone Lemeer
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Christine Dierks
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Peschel
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Internal Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | | | - Jesus Duque-Afonso
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael L. Cleary
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Detlev Schindler
- Department of Human Genetics, Institute of Human Genetics, Biozentrum, University of Würzburg, Würzburg, Germany
| | | | - Melanie Boerries
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, University Medical Center — University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Charlotte M. Niemeyer
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, and
| | - Robert A.J. Oostendorp
- Department of Internal Medicine III, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Justus Duyster
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna Lena Illert
- Department of Internal Medicine I, Medical Center — University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
23
|
Lowran K, Campbell L, Popp P, Wu CG. Assembly of a G-Quadruplex Repair Complex by the FANCJ DNA Helicase and the REV1 Polymerase. Genes (Basel) 2019; 11:E5. [PMID: 31861576 PMCID: PMC7017153 DOI: 10.3390/genes11010005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022] Open
Abstract
The FANCJ helicase unfolds G-quadruplexes (G4s) in human cells to support DNA replication. This action is coupled to the recruitment of REV1 polymerase to synthesize DNA across from a guanine template. The precise mechanisms of these reactions remain unclear. While FANCJ binds to G4s with an AKKQ motif, it is not known whether this site recognizes damaged G4 structures. FANCJ also has a PIP-like (PCNA Interacting Protein) region that may recruit REV1 to G4s either directly or through interactions mediated by PCNA protein. In this work, we measured the affinities of a FANCJ AKKQ peptide for G4s formed by (TTAGGG)4 and (GGGT)4 using fluorescence spectroscopy and biolayer interferometry (BLI). The effects of 8-oxoguanine (8oxoG) on these interactions were tested at different positions. BLI assays were then performed with a FANCJ PIP to examine its recruitment of REV1 and PCNA. FANCJ AKKQ bound tightly to a TTA loop and was sequestered away from the 8oxoG. Reducing the loop length between guanine tetrads increased the affinity of the peptide for 8oxoG4s. FANCJ PIP targeted both REV1 and PCNA but favored interactions with the REV1 polymerase. The impact of these results on the remodeling of damaged G4 DNA is discussed herein.
Collapse
Affiliation(s)
- Kaitlin Lowran
- Department of Chemistry, Oakland University, Rochester, MI 48309, USA; (K.L.); (L.C.)
| | - Laura Campbell
- Department of Chemistry, Oakland University, Rochester, MI 48309, USA; (K.L.); (L.C.)
| | - Phillip Popp
- Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Colin G. Wu
- Department of Chemistry, Oakland University, Rochester, MI 48309, USA; (K.L.); (L.C.)
| |
Collapse
|
24
|
Wu Z, Zhang X, He Z, Hou L. Identifying candidate diagnostic markers for early stage of non-small cell lung cancer. PLoS One 2019; 14:e0225080. [PMID: 31726467 PMCID: PMC6855900 DOI: 10.1371/journal.pone.0225080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 10/28/2019] [Indexed: 11/19/2022] Open
Abstract
We performed a series of bioinformatics analysis on a set of important gene expression data with 76 samples in early stage of non-small cell lung cancer, including 40 adenocarcinoma samples, 16 squamous cell carcinoma samples and 20 normal samples. In order to identify the specific markers for diagnosis, we compared the two subtypes with the normal samples respectively to determine the gene expression characteristics. Through the multi-dimensional scaling classification, we found that the samples were clustered well according to the disease cases. Based on the classification results and using empirical Bayes moderation and treat method, 486 important genes associated with the disease were identified. We constructed gene functions and gene pathways to verify our result and explain the pathogenicity factor and process. We generated a protein-protein interaction network based on the mutual interaction between the selected genes and found that the top thirteen hub genes were highly associated with lung cancer or some other cancers including five newly found genes through our method. The results of this study indicated that contrast on the gene expression between different subtypes and normal samples provides important information for the detection of non-small cell lung cancer and helps exploration of the disease pathogenesis.
Collapse
Affiliation(s)
- Zhen Wu
- School of Mathematics and Statistics, Southwest University, Chongqing 400715, China
| | - Xu Zhang
- School of Mathematics and Statistics, Southwest University, Chongqing 400715, China
| | - Zhihui He
- Department of Pediatric Respiration, Chongqing Ninth People’s Hospital, Chongqing 400700, China
| | - Liyun Hou
- School of Mathematics and Statistics, Southwest University, Chongqing 400715, China
| |
Collapse
|
25
|
Brendel C, Rio P, Verhoeyen E. Humanized mice are precious tools for evaluation of hematopoietic gene therapies and preclinical modeling to move towards a clinical trial. Biochem Pharmacol 2019; 174:113711. [PMID: 31726047 DOI: 10.1016/j.bcp.2019.113711] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/07/2019] [Indexed: 12/11/2022]
Abstract
Over the last decade, incrementally improved xenograft mouse models, which support the engraftment and development of a human hemato-lymphoid system, have been developed and represent an important fundamental and preclinical research tool. Immunodeficient mice can be transplanted with human hematopoietic stem cells (HSCs) and this process is accompanied by HSC homing to the murine bone marrow. This is followed by stem cell expansion, multilineage hematopoiesis, long-term engraftment, and functional human antibody and cellular immune responses. The most significant contributions made by these humanized mice are the identification of normal and leukemic hematopoietic stem cells, the characterization of the human hematopoietic hierarchy, screening of anti-cancer therapies and their use as preclinical models for gene therapy applications. This review article focuses on several gene therapy applications that have benefited from evaluation in humanized mice such as chimeric antigen receptor (CAR) T cell therapies for cancer, anti-viral therapies and gene therapies for multiple monogenetic diseases. Humanized mouse models have been and still are of great value for the gene therapy field since they provide a more reliable understanding of sometimes complicated therapeutic approaches such as recently developed therapeutic gene editing strategies, which seek to correct a gene at its endogenous genomic locus. Additionally, humanized mouse models, which are of great importance with regard to testing new vector technologies in vivo for assessing safety and efficacy prior toclinical trials, help to expedite the critical translation from basic findings to clinical applications. In this review, innovative gene therapies and preclinical studies to evaluate T- and B-cell and HSC-based therapies in humanized mice are discussed and illustrated by multiple examples.
Collapse
Affiliation(s)
- Christian Brendel
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Paula Rio
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Els Verhoeyen
- CIRI, Université de Lyon, INSERM U1111, ENS de Lyon, Université Lyon1, CNRS, UMR 5308, 69007 Lyon, France; Université Côte d'Azur, INSERM, C3M, 06204 Nice, France.
| |
Collapse
|
26
|
Río P, Navarro S, Wang W, Sánchez-Domínguez R, Pujol RM, Segovia JC, Bogliolo M, Merino E, Wu N, Salgado R, Lamana ML, Yañez RM, Casado JA, Giménez Y, Román-Rodríguez FJ, Álvarez L, Alberquilla O, Raimbault A, Guenechea G, Lozano ML, Cerrato L, Hernando M, Gálvez E, Hladun R, Giralt I, Barquinero J, Galy A, García de Andoín N, López R, Catalá A, Schwartz JD, Surrallés J, Soulier J, Schmidt M, Díaz de Heredia C, Sevilla J, Bueren JA. Successful engraftment of gene-corrected hematopoietic stem cells in non-conditioned patients with Fanconi anemia. Nat Med 2019; 25:1396-1401. [PMID: 31501599 DOI: 10.1038/s41591-019-0550-z] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 07/18/2019] [Indexed: 12/20/2022]
Abstract
Fanconi anemia (FA) is a DNA repair syndrome generated by mutations in any of the 22 FA genes discovered to date1,2. Mutations in FANCA account for more than 60% of FA cases worldwide3,4. Clinically, FA is associated with congenital abnormalities and cancer predisposition. However, bone marrow failure is the primary pathological feature of FA that becomes evident in 70-80% of patients with FA during the first decade of life5,6. In this clinical study (ClinicalTrials.gov, NCT03157804 ; European Clinical Trials Database, 2011-006100-12), we demonstrate that lentiviral-mediated hematopoietic gene therapy reproducibly confers engraftment and proliferation advantages of gene-corrected hematopoietic stem cells (HSCs) in non-conditioned patients with FA subtype A. Insertion-site analyses revealed the multipotent nature of corrected HSCs and showed that the repopulation advantage of these cells was not due to genotoxic integrations of the therapeutic provirus. Phenotypic correction of blood and bone marrow cells was shown by the acquired resistance of hematopoietic progenitors and T lymphocytes to DNA cross-linking agents. Additionally, an arrest of bone marrow failure progression was observed in patients with the highest levels of gene marking. The progressive engraftment of corrected HSCs in non-conditioned patients with FA supports that gene therapy should constitute an innovative low-toxicity therapeutic option for this life-threatening disorder.
Collapse
Affiliation(s)
- Paula Río
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Susana Navarro
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Wei Wang
- Division of Translational Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany.,GeneWerk, Heidelberg, Germany
| | - Rebeca Sánchez-Domínguez
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Roser M Pujol
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Servicio de Genética, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Instituto de Investigaciones Biomédicas, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - José C Segovia
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Massimo Bogliolo
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Servicio de Genética, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Instituto de Investigaciones Biomédicas, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Eva Merino
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Servicio de Hematología y Oncología Pediátrica, Fundación de Investigación Biomédica, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Ning Wu
- Division of Translational Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany
| | - Rocío Salgado
- Servicio de Hematología, Hospital Universitario Fundación Jiménez Diaz, Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - María L Lamana
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Rosa M Yañez
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - José A Casado
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Yari Giménez
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Francisco J Román-Rodríguez
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Lara Álvarez
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Omaira Alberquilla
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Anna Raimbault
- Université de Paris (IRSL, INSERM, CNRS), Paris, France.,Hôpital Saint-Louis, Paris, France
| | - Guillermo Guenechea
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - M Luz Lozano
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Laura Cerrato
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Miriam Hernando
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain
| | - Eva Gálvez
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Servicio de Hematología y Oncología Pediátrica, Fundación de Investigación Biomédica, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Raquel Hladun
- Servicio de Oncología y Hematología Pediátricas, Hospital Universitari Vall d'Hebron, Barcelona, Spain.,Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Irina Giralt
- Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | | | | | | | - Ricardo López
- Osakidetza Basque Health Service, Pediatric Oncology and Hematology Unit, Cruces University Hospital, Barakaldo, Spain
| | - Albert Catalá
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Servicio de Hematología y Oncología, Hospital Sant Joan de Déu, Barcelona, Spain
| | | | - Jordi Surrallés
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Servicio de Genética, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Instituto de Investigaciones Biomédicas, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jean Soulier
- Université de Paris (IRSL, INSERM, CNRS), Paris, France.,Hôpital Saint-Louis, Paris, France
| | - Manfred Schmidt
- Division of Translational Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany.,GeneWerk, Heidelberg, Germany
| | - Cristina Díaz de Heredia
- Servicio de Oncología y Hematología Pediátricas, Hospital Universitari Vall d'Hebron, Barcelona, Spain.,Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Julián Sevilla
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.,Servicio de Hematología y Oncología Pediátrica, Fundación de Investigación Biomédica, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Juan A Bueren
- Hematopoietic Innovative Therapies Division, Centro de investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain. .,Instituto de Investigaciones Sanitarias de la Fundación Jiménez Díaz, Madrid, Spain.
| |
Collapse
|
27
|
Tutt A. Inhibited, trapped or adducted: the optimal selective synthetic lethal mix for BRCAness. Ann Oncol 2019; 29:18-21. [PMID: 29300815 PMCID: PMC5834033 DOI: 10.1093/annonc/mdx775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- A Tutt
- Breast Cancer Now Research Centre, Institute of Cancer Research, London, UK.,Research Oncology, Kings College London, London, UK
| |
Collapse
|
28
|
Esteban-Medina M, Peña-Chilet M, Loucera C, Dopazo J. Exploring the druggable space around the Fanconi anemia pathway using machine learning and mechanistic models. BMC Bioinformatics 2019; 20:370. [PMID: 31266445 PMCID: PMC6604281 DOI: 10.1186/s12859-019-2969-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In spite of the abundance of genomic data, predictive models that describe phenotypes as a function of gene expression or mutations are difficult to obtain because they are affected by the curse of dimensionality, given the disbalance between samples and candidate genes. And this is especially dramatic in scenarios in which the availability of samples is difficult, such as the case of rare diseases. RESULTS The application of multi-output regression machine learning methodologies to predict the potential effect of external proteins over the signaling circuits that trigger Fanconi anemia related cell functionalities, inferred with a mechanistic model, allowed us to detect over 20 potential therapeutic targets. CONCLUSIONS The use of artificial intelligence methods for the prediction of potentially causal relationships between proteins of interest and cell activities related with disease-related phenotypes opens promising avenues for the systematic search of new targets in rare diseases.
Collapse
Affiliation(s)
- Marina Esteban-Medina
- Clinical Bioinformatics Area. Fundación Progreso y Salud (FPS). CDCA, Hospital Virgen del Rocio, 41013 Sevilla, Spain
| | - María Peña-Chilet
- Clinical Bioinformatics Area. Fundación Progreso y Salud (FPS). CDCA, Hospital Virgen del Rocio, 41013 Sevilla, Spain
- Bioinformatics in Rare Diseases (BiER). Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocío, 41013 Sevilla, Spain
| | - Carlos Loucera
- Clinical Bioinformatics Area. Fundación Progreso y Salud (FPS). CDCA, Hospital Virgen del Rocio, 41013 Sevilla, Spain
| | - Joaquín Dopazo
- Clinical Bioinformatics Area. Fundación Progreso y Salud (FPS). CDCA, Hospital Virgen del Rocio, 41013 Sevilla, Spain
- Bioinformatics in Rare Diseases (BiER). Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocío, 41013 Sevilla, Spain
- INB-ELIXIR-es, FPS, Hospital Virgen del Rocío, 42013 Sevilla, Spain
| |
Collapse
|
29
|
Faraoni I, Giansanti M, Voso MT, Lo-Coco F, Graziani G. Targeting ADP-ribosylation by PARP inhibitors in acute myeloid leukaemia and related disorders. Biochem Pharmacol 2019; 167:133-148. [PMID: 31028744 DOI: 10.1016/j.bcp.2019.04.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/16/2019] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukaemia (AML) is a highly heterogeneous disease characterized by uncontrolled proliferation, block in myeloid differentiation and recurrent genetic abnormalities. In the search of new effective therapies, identification of synthetic lethal partners of AML genetic alterations might represent a suitable approach to tailor patient treatment. Genetic mutations directly affecting DNA repair genes are not commonly present in AML. Nevertheless, several studies indicate that AML cells show high levels of DNA lesions and genomic instability. Leukaemia-driving oncogenes (e.g., RUNX1-RUNXT1, PML-RARA, TCF3-HLF, IDH1/2, TET2) or treatment with targeted agents directed against aberrant kinases (e.g., JAK1/2 and FLT3 inhibitors) have been associated with reduced DNA repair gene expression/activity that would render leukaemia blasts selectively sensitive to synthetic lethality induced by poly(ADP-ribose) polymerase inhibitors (PARPi). Thus, specific oncogenic chimeric proteins or gene mutations, rare or typically distinctive of certain leukaemia subtypes, may allow tagging cancer cells for destruction by PARPi. In this review, we will discuss the rationale for using PARPi in AML subtypes characterized by a specific genetic background and summarize the preclinical and clinical evidence reported so far on their activity when used as single agents or in combination with classical cytotoxic chemotherapy or with agents targeting AML-associated mutated proteins.
Collapse
Affiliation(s)
- Isabella Faraoni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Manuela Giansanti
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Francesco Lo-Coco
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy; Unit of Neuro-Oncohematology, Santa Lucia Foundation-I.R.C.C.S., Rome, Italy
| | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
30
|
Onishi S, Tajika M, Tanaka T, Ishihara M, Hirayama Y, Mizuno N, Kuwahara T, Okuno N, Matsumoto S, Toriyama K, Kurita Y, Obata M, Koide Y, Hasegawa Y, Hara K, Niwa Y. Superficial Esophageal Cancer in a Fanconi Anemia Patient That Was Treated Successfully by Endoscopic Submucosal Resection. Intern Med 2019; 58:529-533. [PMID: 30333403 PMCID: PMC6421153 DOI: 10.2169/internalmedicine.1434-18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/02/2018] [Indexed: 11/06/2022] Open
Abstract
Fanconi anemia (FA) is a disorder of chromosomal fragility characterized by progression to aplastic anemia, myelodysplastic syndrome, and leukemia. FA patients are also predisposed to solid cancers. A case of FA in an adult patient who developed tongue and superficial esophageal cancers following hematopoietic stem cell transplantation is reported. This case was considered significant because it is the first reported case of superficial esophageal cancer in an FA patient that was treated successfully by endoscopic submucosal resection.
Collapse
Affiliation(s)
- Sachiyo Onishi
- Department of Endoscopy, Aichi Cancer Center Hospital, Japan
| | - Masahiro Tajika
- Department of Endoscopy, Aichi Cancer Center Hospital, Japan
| | - Tsutomu Tanaka
- Department of Endoscopy, Aichi Cancer Center Hospital, Japan
| | - Makoto Ishihara
- Department of Endoscopy, Aichi Cancer Center Hospital, Japan
| | - Yutaka Hirayama
- Department of Endoscopy, Aichi Cancer Center Hospital, Japan
| | - Nobumasa Mizuno
- Department of Gastroenterology, Aichi Cancer Center Hospital, Japan
| | | | - Nozomi Okuno
- Department of Gastroenterology, Aichi Cancer Center Hospital, Japan
| | | | | | - Yusuke Kurita
- Department of Gastroenterology, Aichi Cancer Center Hospital, Japan
| | - Masanori Obata
- Department of Gastroenterology, Aichi Cancer Center Hospital, Japan
| | - Yusuke Koide
- Department of Head and Neck Surgery, Aichi Cancer Center Hospital, Japan
| | - Yasuhisa Hasegawa
- Department of Head and Neck Surgery, Aichi Cancer Center Hospital, Japan
| | - Kazuo Hara
- Department of Gastroenterology, Aichi Cancer Center Hospital, Japan
| | - Yasumasa Niwa
- Department of Endoscopy, Aichi Cancer Center Hospital, Japan
| |
Collapse
|
31
|
M. AlDallal S. Quick glance at Fanconi anemia and BRCA2/FANCD1. AIMS MEDICAL SCIENCE 2019. [DOI: 10.3934/medsci.2019.4.326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
32
|
Velmurugan KR, Michalak P, Kang L, Fonville NC, Garner HR. Dysfunctional DNA repair pathway via defective FANCD2 gene engenders multifarious exomic and transcriptomic effects in Fanconi anemia. Mol Genet Genomic Med 2018; 6:1199-1208. [PMID: 30450770 PMCID: PMC6305641 DOI: 10.1002/mgg3.502] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/20/2018] [Accepted: 10/10/2018] [Indexed: 01/27/2023] Open
Abstract
Background Fanconi anemia (FA) affects only one in 130,000 births, but has severe and diverse clinical consequences. It has been theorized that defects in the FA DNA cross‐link repair complex lead to a spectrum of variants that are responsible for those diverse clinical phenotypes. Methods Using NextGen sequencing, we show that a clinically derived FA cell line had accumulated numerous genetic variants, including high‐impact mutations, such as deletion of start codons, introduction of premature stop codons, missense mutations, and INDELs. Results About 65% of SNPs and 55% of INDELs were found to be commonly present in both the FA dysfunctional and retrovirally corrected cell lines, showing their common origin. The number of INDELs, but not SNPs, is decreased in FANCD2‐corrected samples, suggesting that FANCD2 deficiency preferentially promotes the origin of INDELs. These genetic modifications had a considerable effect on the transcriptome, with statistically significant changes in the expression of 270 genes. These genetic and transcriptomic variants significantly impacted pathways and molecular functions, spanning a diverse spectrum of disease phenotypes/symptoms, consistent with the disease diversity seen in FA patients. Conclusion These results underscore the consequences of defects in the DNA cross‐link repair mechanism and indicate that accumulating diverse mutations from individual parent cells may make it difficult to anticipate the longitudinal clinical behavior of emerging disease states in an individual with FA.
Collapse
Affiliation(s)
- Karthik Raja Velmurugan
- Primary Care Research Network and the Center for Bioinformatics and Genetics, Edward Via College of Osteopathic Medicine, Blacksburg, Virginia
| | - Pawel Michalak
- Primary Care Research Network and the Center for Bioinformatics and Genetics, Edward Via College of Osteopathic Medicine, Blacksburg, Virginia.,Center for One Health Research, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia.,Institute of Evolution, University of Haifa, Haifa, Israel
| | - Lin Kang
- Primary Care Research Network and the Center for Bioinformatics and Genetics, Edward Via College of Osteopathic Medicine, Blacksburg, Virginia
| | | | - Harold R Garner
- Primary Care Research Network and the Center for Bioinformatics and Genetics, Edward Via College of Osteopathic Medicine, Blacksburg, Virginia.,The Gibbs Cancer Center and Research Institute, Spartanburg, South Carolina
| |
Collapse
|
33
|
Kim H, Schaniel C. Modeling Hematological Diseases and Cancer With Patient-Specific Induced Pluripotent Stem Cells. Front Immunol 2018; 9:2243. [PMID: 30323816 PMCID: PMC6172418 DOI: 10.3389/fimmu.2018.02243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/10/2018] [Indexed: 12/13/2022] Open
Abstract
The advent of induced pluripotent stem cells (iPSCs) together with recent advances in genome editing, microphysiological systems, tissue engineering and xenograft models present new opportunities for the investigation of hematological diseases and cancer in a patient-specific context. Here we review the progress in the field and discuss the advantages, limitations, and challenges of iPSC-based malignancy modeling. We will also discuss the use of iPSCs and its derivatives as cellular sources for drug target identification, drug development and evaluation of pharmacological responses.
Collapse
Affiliation(s)
- Huensuk Kim
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Christoph Schaniel
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
34
|
Weyand CM, Shen Y, Goronzy JJ. Redox-sensitive signaling in inflammatory T cells and in autoimmune disease. Free Radic Biol Med 2018; 125:36-43. [PMID: 29524605 PMCID: PMC6128787 DOI: 10.1016/j.freeradbiomed.2018.03.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 03/01/2018] [Accepted: 03/04/2018] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) are byproducts of oxygen metabolism best known for their damaging potential, but recent evidence has exposed their role as secondary messengers, which regulate cell function through redox-activatable signaling systems. In immune cells, specifically in T cells, redox-sensitive signaling pathways have been implicated in controlling several functional domains; including cell cycle progression, T effector cell differentiation, tissue invasion and inflammatory behavior. T cells from patients with the autoimmune disease rheumatoid arthritis (RA) have emerged as a valuable model system to examine the functional impact of ROS on T cell function. Notably, RA T cells are distinguished from healthy T cells based on reduced ROS production and undergo "reductive stress". Upstream defects leading to the ROSlow status of RA T cells are connected to metabolic reorganization. RA T cells shunt glucose away from pyruvate and ATP production towards the pentose phosphate pathway, where they generate NADPH and consume cellular ROS. Downstream consequences of the ROSlow conditions in RA T cells include insufficient activation of the DNA repair kinase ATM, bypassing of the G2/M cell cycle checkpoint and biased differentiation of T cells into IFN-γ and IL-17-producing inflammatory cells. Also, ROSlow T cells rapidly invade into peripheral tissue due to dysregulated lipogenesis, excessive membrane ruffling, and overexpression of a motility module dominated by the scaffolding protein Tks5. These data place ROS into a pinnacle position in connecting cellular metabolism and protective versus auto-aggressive T cell immunity. Therapeutic interventions for targeted ROS enhancement instead of ROS depletion should be developed as a novel strategy to treat autoimmune tissue inflammation.
Collapse
Affiliation(s)
- Cornelia M Weyand
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Veterans Affairs Palo Alto Health Care System Palo Alto, CA 94306, USA.
| | - Yi Shen
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Veterans Affairs Palo Alto Health Care System Palo Alto, CA 94306, USA
| | - Jorg J Goronzy
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Veterans Affairs Palo Alto Health Care System Palo Alto, CA 94306, USA
| |
Collapse
|
35
|
Ji K, Kong Y, Liu Y, Wang Y, Du L, Xu C, Li Y, Zhang F, Zhu X, Liu Q. Evaluation on the genetic instability detecting methods for rapid diagnose of Fanconi anemia used in the undeveloped areas of China. Int J Lab Hematol 2018; 40:630-636. [DOI: 10.1111/ijlh.12878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 05/08/2018] [Indexed: 11/28/2022]
Affiliation(s)
- Kaihua Ji
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine; Department of Radiobiology; Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College; Tianjin China
| | - Yangyang Kong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine; Department of Radiobiology; Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College; Tianjin China
| | - Yang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine; Department of Radiobiology; Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College; Tianjin China
| | - Yan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine; Department of Radiobiology; Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College; Tianjin China
| | - Liqing Du
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine; Department of Radiobiology; Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College; Tianjin China
| | - Chang Xu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine; Department of Radiobiology; Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College; Tianjin China
| | - Yuan Li
- Chinese Academy of Medical Science Therapeutic Center of Anemia; Institute of Hematology & Blood Diseases Hospital; Chinese Academy of Medical Science & Pecking Union Medical College; Tianjin China
| | - Fengkui Zhang
- Chinese Academy of Medical Science Therapeutic Center of Anemia; Institute of Hematology & Blood Diseases Hospital; Chinese Academy of Medical Science & Pecking Union Medical College; Tianjin China
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, Diagnosis and Treatment Center of Pediatric Blood Diseases; Institute of Hematology and Blood Diseases Hospital; Center for Stem Cell Medicine; Chinese Academy of Medical Sciences & Peking Union Medical College; Tianjin China
| | - Qiang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine; Department of Radiobiology; Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College; Tianjin China
| |
Collapse
|
36
|
Tutt A, Tovey H, Cheang MCU, Kernaghan S, Kilburn L, Gazinska P, Owen J, Abraham J, Barrett S, Barrett-Lee P, Brown R, Chan S, Dowsett M, Flanagan JM, Fox L, Grigoriadis A, Gutin A, Harper-Wynne C, Hatton MQ, Hoadley KA, Parikh J, Parker P, Perou CM, Roylance R, Shah V, Shaw A, Smith IE, Timms KM, Wardley AM, Wilson G, Gillett C, Lanchbury JS, Ashworth A, Rahman N, Harries M, Ellis P, Pinder SE, Bliss JM. Carboplatin in BRCA1/2-mutated and triple-negative breast cancer BRCAness subgroups: the TNT Trial. Nat Med 2018; 24:628-637. [PMID: 29713086 PMCID: PMC6372067 DOI: 10.1038/s41591-018-0009-7] [Citation(s) in RCA: 633] [Impact Index Per Article: 90.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/02/2018] [Indexed: 12/25/2022]
Abstract
Germline mutations in BRCA1/2 predispose individuals to breast cancer (termed germline-mutated BRCA1/2 breast cancer, gBRCA-BC) by impairing homologous recombination (HR) and causing genomic instability. HR also repairs DNA lesions caused by platinum agents and PARP inhibitors. Triple-negative breast cancers (TNBCs) harbor subpopulations with BRCA1/2 mutations, hypothesized to be especially platinum-sensitive. Cancers in putative 'BRCAness' subgroups-tumors with BRCA1 methylation; low levels of BRCA1 mRNA (BRCA1 mRNA-low); or mutational signatures for HR deficiency and those with basal phenotypes-may also be sensitive to platinum. We assessed the efficacy of carboplatin and another mechanistically distinct therapy, docetaxel, in a phase 3 trial in subjects with unselected advanced TNBC. A prespecified protocol enabled biomarker-treatment interaction analyses in gBRCA-BC and BRCAness subgroups. The primary endpoint was objective response rate (ORR). In the unselected population (376 subjects; 188 carboplatin, 188 docetaxel), carboplatin was not more active than docetaxel (ORR, 31.4% versus 34.0%, respectively; P = 0.66). In contrast, in subjects with gBRCA-BC, carboplatin had double the ORR of docetaxel (68% versus 33%, respectively; biomarker, treatment interaction P = 0.01). Such benefit was not observed for subjects with BRCA1 methylation, BRCA1 mRNA-low tumors or a high score in a Myriad HRD assay. Significant interaction between treatment and the basal-like subtype was driven by high docetaxel response in the nonbasal subgroup. We conclude that patients with advanced TNBC benefit from characterization of BRCA1/2 mutations, but not BRCA1 methylation or Myriad HRD analyses, to inform choices on platinum-based chemotherapy. Additionally, gene expression analysis of basal-like cancers may also influence treatment selection.
Collapse
Affiliation(s)
- Andrew Tutt
- Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK.
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital, London, UK.
| | - Holly Tovey
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Maggie Chon U Cheang
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Sarah Kernaghan
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Lucy Kilburn
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Patrycja Gazinska
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital, London, UK
| | - Julie Owen
- King's Health Partners Cancer Biobank, King's College London, London, UK
| | | | | | | | - Robert Brown
- Department of Surgery and Cancer, Imperial College London, London, UK
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Stephen Chan
- Department of Clinical Oncology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Mitchell Dowsett
- Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - James M Flanagan
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Lisa Fox
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Anita Grigoriadis
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital, London, UK
| | | | | | - Matthew Q Hatton
- Department of Clinical Oncology, Weston Park Hospital, Sheffield, UK
| | - Katherine A Hoadley
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jyoti Parikh
- Department of Radiology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, UK
| | - Peter Parker
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Medical School Campus, London, UK
- Protein Phosphorylation Laboratory, Francis Crick Institute, London, UK
| | - Charles M Perou
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rebecca Roylance
- Department of Oncology, University College London Hospitals NHS Foundation Trust and NIHR University College London Hospitals Biomedical Research Centre, London, UK
| | - Vandna Shah
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, Guy's Hospital, London, UK
| | - Adam Shaw
- Department of Medical and Molecular Genetics, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Ian E Smith
- Breast Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | | | - Andrew M Wardley
- NIHR Manchester Clinical Research Facility at The Christie and Division of Cancer Sciences and University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | | | - Cheryl Gillett
- King's Health Partners Cancer Biobank, King's College London, London, UK
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, London, UK
| | | | - Alan Ashworth
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, USA
| | - Nazneen Rahman
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Cancer Genetics Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Mark Harries
- Department of Medical Oncology, Guy's and St Thomas Foundation Trust, London, UK
| | - Paul Ellis
- Department of Medical Oncology, Guy's and St Thomas Foundation Trust, London, UK
| | - Sarah E Pinder
- King's Health Partners Cancer Biobank, King's College London, London, UK
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, London, UK
| | - Judith M Bliss
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| |
Collapse
|
37
|
Che R, Zhang J, Nepal M, Han B, Fei P. Multifaceted Fanconi Anemia Signaling. Trends Genet 2018; 34:171-183. [PMID: 29254745 PMCID: PMC5858900 DOI: 10.1016/j.tig.2017.11.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/28/2017] [Indexed: 01/26/2023]
Abstract
In 1927 Guido Fanconi described a hereditary condition presenting panmyelopathy accompanied by short stature and hyperpigmentation, now better known as Fanconi anemia (FA). With this discovery the genetic and molecular basis underlying FA has emerged as a field of great interest. FA signaling is crucial in the DNA damage response (DDR) to mediate the repair of damaged DNA. This has attracted a diverse range of investigators, especially those interested in aging and cancer. However, recent evidence suggests FA signaling also regulates functions outside the DDR, with implications for many other frontiers of research. We discuss here the characteristics of FA functions and expand upon current perspectives regarding the genetics of FA, indicating that FA plays a role in a myriad of molecular and cellular processes.
Collapse
Affiliation(s)
- Raymond Che
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA; Graduate Program of Molecular Biosciences and Bioengineering, University of Hawaii, Honolulu, HI, USA
| | - Jun Zhang
- Department of Laboratory Medicine and Pathology, Mayo Clinic Foundation, USA
| | - Manoj Nepal
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA; Graduate Program of Molecular Biosciences and Bioengineering, University of Hawaii, Honolulu, HI, USA
| | - Bing Han
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Peiwen Fei
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA; Graduate Program of Molecular Biosciences and Bioengineering, University of Hawaii, Honolulu, HI, USA.
| |
Collapse
|
38
|
Angel J, DiGiovanni J. Genetic Determinants of Cancer Susceptibility. COMPREHENSIVE TOXICOLOGY 2018:330-360. [DOI: 10.1016/b978-0-12-801238-3.65251-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
39
|
Panneerselvam J, Wang H, Zhang J, Che R, Yu H, Fei P. BLM promotes the activation of Fanconi Anemia signaling pathway. Oncotarget 2017; 7:32351-61. [PMID: 27083049 PMCID: PMC5078018 DOI: 10.18632/oncotarget.8707] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 03/28/2016] [Indexed: 12/21/2022] Open
Abstract
Mutations in the human RecQ helicase, BLM, causes Bloom Syndrome, which is a rare autosomal recessive disorder and characterized by genomic instability and an increased risk of cancer. Fanconi Anemia (FA), resulting from mutations in any of the 19 known FA genes and those yet to be known, is also characterized by chromosomal instability and a high incidence of cancer. BLM helicase and FA proteins, therefore, may work in a common tumor-suppressor signaling pathway. To date, it remains largely unclear as to how BLM and FA proteins work concurrently in the maintenance of genome stability. Here we report that BLM is involved in the early activation of FA group D2 protein (FANCD2). We found that FANCD2 activation is substantially delayed and attenuated in crosslinking agent-treated cells harboring deficient Blm compared to similarly treated control cells with sufficient BLM. We also identified that the domain VI of BLM plays an essential role in promoting FANCD2 activation in cells treated with DNA crosslinking agents, especially ultraviolet B. The similar biological effects performed by ΔVI-BLM and inactivated FANCD2 further confirm the relationship between BLM and FANCD2. Mutations within the domain VI of BLM detected in human cancer samples demonstrate the functional importance of this domain, suggesting human tumorigenicity resulting from mtBLM may be at least partly attributed to mitigated FANCD2 activation. Collectively, our data show a previously unknown regulatory liaison in advancing our understanding of how the cancer susceptibility gene products act in concert to maintain genome stability.
Collapse
Affiliation(s)
| | - Hong Wang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.,Current address: Sun Yat-Sen University, Guangzhou, China
| | - Jun Zhang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Raymond Che
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Herbert Yu
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Peiwen Fei
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| |
Collapse
|
40
|
Overlooked FANCD2 variant encodes a promising, portent tumor suppressor, and alternative polyadenylation contributes to its expression. Oncotarget 2017; 8:22490-22500. [PMID: 28157704 PMCID: PMC5410239 DOI: 10.18632/oncotarget.14989] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/24/2017] [Indexed: 01/02/2023] Open
Abstract
Fanconi Anemia (FA) complementation group D2 protein (FANCD2) is the center of the FA tumor suppressor pathway, which has become an important field of investigation in human aging and cancer. Here we report an overlooked central player in the FA pathway, FANCD2 variant 2 (FANCD2-V2), which appears to perform more potent tumor suppressor-function compared to the known variant of FANCD2, namely, FANCD2-V1. Detailed analysis of the FANCD2 gene structure indicated a proximal and distal polyadenylation site (PAS), associated with V2 and V1 transcripts accordingly. RNA polymerase II Chromatin immunoprecipitation (ChIP) targeting the two PAS-regions determined lesser binding of RNA pol II to DNA fragments in the distal PAS region in non-malignant cells compared to malignant cells. Conversely, the opposite occurred in the proximal PAS region. Moreover, RNA immunoprecipitation (RIP) identified that U2 snRNP, a major component of RNA splicing complex that interacts with the 3′end of an intron, showed greater binding to the last intron of the FANCD2-V1 transcript in malignant cells compared to the non-malignant cells. Importantly, our data showed that in human tissue samples, the ratio of V2 /V1 expression in lung, bladder, or ovarian cancer correlates inversely with the tumor stages/grades. Therefore, these findings provide a previously unrecognized central player FANCD2-V2 and thus novel insights into human tumorigenesis, and indicate that V2/V1 can act as an effective biomarker in assisting the recognition of tumor malignance.
Collapse
|
41
|
Nepomuceno TC, De Gregoriis G, de Oliveira FMB, Suarez-Kurtz G, Monteiro AN, Carvalho MA. The Role of PALB2 in the DNA Damage Response and Cancer Predisposition. Int J Mol Sci 2017; 18:ijms18091886. [PMID: 28858227 PMCID: PMC5618535 DOI: 10.3390/ijms18091886] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/21/2017] [Accepted: 08/26/2017] [Indexed: 01/01/2023] Open
Abstract
The deoxyribonucleic acid (DNA) damage response (DDR) is a major feature in the maintenance of genome integrity and in the suppression of tumorigenesis. PALB2 (Partner and Localizer of Breast Cancer 2 (BRCA2)) plays an important role in maintaining genome integrity through its role in the Fanconi anemia (FA) and homologous recombination (HR) DNA repair pathways. Since its identification as a BRCA2 interacting partner, PALB2 has emerged as a pivotal tumor suppressor protein associated to hereditary cancer susceptibility to breast and pancreatic cancers. In this review, we discuss how other DDR proteins (such as the kinases Ataxia Telangiectasia Mutated (ATM) and ATM- and Rad3-Related (ATR), mediators BRCA1 (Breast Cancer 1)/BRCA2 and effectors RAD51/DNA Polymerase η (Polη) interact with PALB2 to orchestrate DNA repair. We also examine the involvement of PALB2 mutations in the predisposition to cancer and the role of PALB2 in stimulating error-free DNA repair through the FA/HR pathway.
Collapse
Affiliation(s)
- Thales C Nepomuceno
- Programa de Pesquisa Clínica, Instituto Nacional de Câncer, Rio de Janeiro 20231-050, Brazil.
| | - Giuliana De Gregoriis
- Programa de Pesquisa Clínica, Instituto Nacional de Câncer, Rio de Janeiro 20231-050, Brazil.
| | | | - Guilherme Suarez-Kurtz
- Programa de Pesquisa Clínica, Instituto Nacional de Câncer, Rio de Janeiro 20231-050, Brazil.
| | - Alvaro N Monteiro
- Cancer Epidemiology Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | - Marcelo A Carvalho
- Programa de Pesquisa Clínica, Instituto Nacional de Câncer, Rio de Janeiro 20231-050, Brazil.
- Instituto Federal do Rio de Janeiro-IFRJ, Rio de Janeiro 20270-021, Brazil.
| |
Collapse
|
42
|
Nepal M, Che R, Ma C, Zhang J, Fei P. FANCD2 and DNA Damage. Int J Mol Sci 2017; 18:ijms18081804. [PMID: 28825622 PMCID: PMC5578191 DOI: 10.3390/ijms18081804] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/08/2017] [Accepted: 08/12/2017] [Indexed: 02/07/2023] Open
Abstract
Investigators have dedicated considerable effort to understanding the molecular basis underlying Fanconi Anemia (FA), a rare human genetic disease featuring an extremely high incidence of cancer and many congenital defects. Among those studies, FA group D2 protein (FANCD2) has emerged as the focal point of FA signaling and plays crucial roles in multiple aspects of cellular life, especially in the cellular responses to DNA damage. Here, we discuss the recent and relevant studies to provide an updated review on the roles of FANCD2 in the DNA damage response.
Collapse
Affiliation(s)
- Manoj Nepal
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA.
- Graduate Program of Molecular Biosciences and Bioengineering, University of Hawaii, Honolulu, HI 96813, USA.
| | - Raymond Che
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA.
- Graduate Program of Molecular Biosciences and Bioengineering, University of Hawaii, Honolulu, HI 96813, USA.
| | - Chi Ma
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA.
| | - Jun Zhang
- Department of Laboratory Medicine and Pathology, Mayo Clinic Foundation, Rochester, MN 55905, USA.
| | - Peiwen Fei
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA.
- Graduate Program of Molecular Biosciences and Bioengineering, University of Hawaii, Honolulu, HI 96813, USA.
| |
Collapse
|
43
|
Involvement of FANCD2 in Energy Metabolism via ATP5α. Sci Rep 2017; 7:4921. [PMID: 28687786 PMCID: PMC5501830 DOI: 10.1038/s41598-017-05150-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 05/24/2017] [Indexed: 12/31/2022] Open
Abstract
Growing evidence supports a general hypothesis that aging and cancer are diseases related to energy metabolism. However, the involvement of Fanconi Anemia (FA) signaling, a unique genetic model system for studying human aging or cancer, in energy metabolism remains elusive. Here, we report that FA complementation group D2 protein (FANCD2) functionally impacts mitochondrial ATP production through its interaction with ATP5α, whereas this relationship was not observed in the mutant FANCD2 (K561R)-carrying cells. Moreover, while ATP5α is present within the mitochondria in wild-type cells, it is instead located mostly outside in cells that carry the non-monoubiquitinated FANCD2. In addition, mitochondrial ATP production is significantly reduced in these cells, compared to those cells carrying wtFANCD2. We identified one region (AA42-72) of ATP5α, contributing to the interaction between ATP5α and FANCD2, which was confirmed by protein docking analysis. Further, we demonstrated that mtATP5α (∆AA42-72) showed an aberrant localization, and resulted in a decreased ATP production, similar to what was observed in non-monoubiquitinated FANCD2-carrying cells. Collectively, our study demonstrates a novel role of FANCD2 in governing cellular ATP production, and advances our understanding of how defective FA signaling contributes to aging and cancer at the energy metabolism level.
Collapse
|
44
|
Aymun U, Iram S, Aftab I, Khaliq S, Nadir A, Nisar A, Mohsin S. Screening for mutations in two exons of FANCG gene in Pakistani population. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2017. [PMID: 28627524 DOI: 10.5507/bp.2017.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Fanconi anemia is a rare autosomal recessive disorder of genetic instability. It is both molecularly and clinically, a heterogeneous disorder. Its incidence is 1 in 129,000 births and relatively high in some ethnic groups. Sixteen genes have been identified among them mutations in FANCG gene are most common after FANCA and FANCC gene mutations. OBJECTIVE To study mutations in exon 3 and 4 of FANCG gene in Pakistani population. METHODS Thirty five patients with positive Diepoxybutane test were included in the study. DNA was extracted and amplified for exons 3 and 4. Thereafter Sequencing was done and analyzed for the presence of mutations. RESULTS No mutation was detected in exon 3 whereas a carrier of known mutation c.307+1 G>T was found in exon 4 of the FANCG gene. CONCLUSION Absence of any mutation in exon 3 and only one heterozygous mutation in exon 4 of FANCG gene points to a different spectrum of FA gene pool in Pakistan that needs extensive research in this area.
Collapse
Affiliation(s)
- Ujala Aymun
- Department of Hematology, University of Health Sciences, Lahore, Pakistan.,Department of Pathology, Avicenna Medical College, Lahore, Pakistan
| | - Saima Iram
- Department of Hematology, University of Health Sciences, Lahore, Pakistan.,Department of Pathology, Bolan Medical College, Quetta, Pakistan
| | - Iram Aftab
- Department of Hematology, University of Health Sciences, Lahore, Pakistan
| | - Saba Khaliq
- Department of Physiology and Cell Biology, University of Health Sciences, Lahore, Pakistan
| | - Ali Nadir
- Department of Hematology, Armed Forces institute of Pathology, Rawalpindi, Pakistan
| | - Ahmed Nisar
- Department of Hematology, Children Hospital Lahore, Pakistan
| | - Shahida Mohsin
- Department of Hematology, University of Health Sciences, Lahore, Pakistan
| |
Collapse
|
45
|
Moghadam AAS, Mahjoubi F, Reisi N, Vosough P. Investigation of FANCA gene in Fanconi anaemia patients in Iran. Indian J Med Res 2017; 143:184-96. [PMID: 27121516 PMCID: PMC4859127 DOI: 10.4103/0971-5916.180206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND & OBJECTIVES Fanconi anaemia (FA) is a syndrome with a predisposition to bone marrow failure, congenital anomalies and malignancies. It is characterized by cellular hypersensitivity to cross-linking agents such as mitomycin C (MMC). In the present study, a new approach was selected to investigate FANCA (Fanconi anaemia complementation group A) gene in patients clinically diagnosed with cellular hypersensitivity to DNA cross-linking agent MMC. METHODS Chromosomal breakage analysis was performed to prove the diagnosis of Fanconi anaemia in 318 families. Of these, 70 families had a positive result. Forty families agreed to molecular genetic testing. In total, there were 27 patients with unknown complementary types. Genomic DNA was extracted and total RNA was isolated from fresh whole blood of the patients. The first-strand cDNA was synthesized and the cDNA of each patient was then tested with 21 pairs of overlapping primers. High resolution melting curve analysis was used to screen FANCA, and LinReg software version 1.7 was utilized for analysis of expression. RESULTS In total, six sequence alterations were identified, which included two stop codons, two frames-shift mutations, one large deletion and one amino acid exchange. FANCA expression was downregulated in patients who had sequence alterations. INTERPRETATION & CONCLUSIONS The results of the present study show that high resolution melting (HRM) curve analysis may be useful in the detection of sequence alteration. It is simpler and more cost-effective than the multiplex ligation-dependent probe amplification (MLPA) procedure.
Collapse
Affiliation(s)
| | - Frouzandeh Mahjoubi
- Medical Biotechnology Institute, National Institute of Genetic Engineering & Biotechnology (NIGEB), Tehran, Iran
| | | | | |
Collapse
|
46
|
Sekinaka Y, Mitsuiki N, Imai K, Yabe M, Yabe H, Mitsui-Sekinaka K, Honma K, Takagi M, Arai A, Yoshida K, Okuno Y, Shiraishi Y, Chiba K, Tanaka H, Miyano S, Muramatsu H, Kojima S, Hira A, Takata M, Ohara O, Ogawa S, Morio T, Nonoyama S. Common Variable Immunodeficiency Caused by FANC Mutations. J Clin Immunol 2017; 37:434-444. [PMID: 28493158 DOI: 10.1007/s10875-017-0396-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 04/17/2017] [Indexed: 11/24/2022]
Abstract
Common variable immunodeficiency (CVID) is the most common adult-onset primary antibody deficiency disease due to various causative genes. Several genes, which are known to be the cause of different diseases, have recently been reported as the cause of CVID in patients by performing whole exome sequencing (WES) analysis. Here, we found FANC gene mutations as a cause of adult-onset CVID in two patients. B cells were absent and CD4+ T cells were skewed toward CD45RO+ memory T cells. T-cell receptor excision circles (TRECs) and signal joint kappa-deleting recombination excision circles (sjKRECs) were undetectable in both patients. Both patients had no anemia, neutropenia, or thrombocytopenia. Using WES, we identified compound heterozygous mutations of FANCE in one patient and homozygous mutation of FANCA in another patient. The impaired function of FANC protein complex was confirmed by a monoubiquitination assay and by chromosome fragility test. We then performed several immunological evaluations including quantitative lymphocyte analysis and TRECs/sjKRECs analysis for 32 individuals with Fanconi anemia (FA). In total, 22 FA patients (68.8%) were found to have immunological abnormalities, suggesting that such immunological findings may be common in FA patients. These data indicate that FANC mutations are involved in impaired lymphogenesis probably by the accumulation of DNA replication stress, leading to CVID. It is important to diagnose FA because it drastically changes clinical management. We propose that FANC mutations can cause isolated immunodeficiency in addition to bone marrow failure and malignancy.
Collapse
Affiliation(s)
- Yujin Sekinaka
- Department of Pediatrics, National Defense Medical College, Saitama, Japan
| | - Noriko Mitsuiki
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kohsuke Imai
- Department of Pediatrics, National Defense Medical College, Saitama, Japan. .,Department of Community Pediatrics, Perinatal and Maternal Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.
| | - Miharu Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Hiromasa Yabe
- Department of Cell Transplantation and Regenerative Medicine, Tokai University School of Medicine, Isehara, Japan
| | | | - Kenichi Honma
- Department of Pediatrics, National Defense Medical College, Saitama, Japan
| | - Masatoshi Takagi
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ayako Arai
- Department of Hematology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenichi Yoshida
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yusuke Okuno
- Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichi Shiraishi
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kenichi Chiba
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroko Tanaka
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoru Miyano
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hideki Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seiji Kojima
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Asuka Hira
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Kyoto University, Kyoto, Japan
| | - Minoru Takata
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Kyoto University, Kyoto, Japan
| | - Osamu Ohara
- Department of Technology Development, Kazusa DNA Research Institute, Kisarazu, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Cancer Genomics Project, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigeaki Nonoyama
- Department of Pediatrics, National Defense Medical College, Saitama, Japan
| |
Collapse
|
47
|
Targeting the ATR-CHK1 Axis in Cancer Therapy. Cancers (Basel) 2017; 9:cancers9050041. [PMID: 28448462 PMCID: PMC5447951 DOI: 10.3390/cancers9050041] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/23/2017] [Accepted: 04/25/2017] [Indexed: 12/14/2022] Open
Abstract
Targeting the DNA damage response (DDR) is a new therapeutic approach in cancer that shows great promise for tumour selectivity. Key components of the DDR are the ataxia telangiectasia mutated and Rad3 related (ATR) and checkpoint kinase 1 (CHK1) kinases. This review article describes the role of ATR and its major downstream target, CHK1, in the DDR and why cancer cells are particularly reliant on the ATR-CHK1 pathway, providing the rationale for targeting these kinases, and validation of this hypothesis by genetic manipulation. The recent development of specific inhibitors and preclinical data using these inhibitors not only as chemosensitisers and radiosensitisers but also as single agents to exploit specific pathologies of tumour cells is described. These potent and specific inhibitors have now entered clinical trial and early results are presented.
Collapse
|
48
|
Sertorio M, Du W, Amarachintha S, Wilson A, Pang Q. In Vivo RNAi Screen Unveils PPARγ as a Regulator of Hematopoietic Stem Cell Homeostasis. Stem Cell Reports 2017; 8:1242-1255. [PMID: 28416286 PMCID: PMC5425620 DOI: 10.1016/j.stemcr.2017.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 03/09/2017] [Accepted: 03/10/2017] [Indexed: 12/13/2022] Open
Abstract
Hematopoietic stem cell (HSC) defects can cause repopulating impairment leading to hematologic diseases. To target HSC deficiency in a disease setting, we exploited the repopulating defect of Fanconi anemia (FA) HSCs to conduct an in vivo short hairpin RNA (shRNA) screen. We exposed Fancd2−/− HSCs to a lentiviral shRNA library targeting 947 genes. We found enrichment of shRNAs targeting genes involved in the PPARγ pathway that has not been linked to HSC homeostasis. PPARγ inhibition by shRNA or chemical compounds significantly improves the repopulating ability of Fancd2−/− HSCs. Conversely, activation of PPARγ in wild-type HSCs impaired hematopoietic repopulation. In mouse HSCs and patient-derived lymphoblasts, PPARγ activation is manifested in upregulating the p53 target p21. PPARγ and co-activators are upregulated in total bone marrow and stem/progenitor cells from FA patients. Collectively, this work illustrates the utility of RNAi technology coupled with HSC transplantation for the discovery of novel genes and pathways involved in stress hematopoiesis. In vivo screening identifies of deleterious Pparγ effect on HSCs Pharmacological activation of Pparγ impaired normal HSC repopulation Inhibition of Pparγ improves Fancd2-deficient HSC repopulation ability
Collapse
Affiliation(s)
- Mathieu Sertorio
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Wei Du
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Surya Amarachintha
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Andrew Wilson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Qishen Pang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
49
|
Impairment of fetal hematopoietic stem cell function in the absence of Fancd2. Exp Hematol 2016; 48:79-86. [PMID: 27915139 DOI: 10.1016/j.exphem.2016.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/23/2016] [Accepted: 11/24/2016] [Indexed: 12/12/2022]
Abstract
Fanconi anemia (FA) results from mutations in the genes necessary for DNA damage repair and often leads to progressive bone marrow failure. Although the exhaustion of the bone marrow leads to cytopenias in FA patients as they age, evidence from human FA and mouse model fetal livers suggests that hematopoietic defects originate in utero, which may lead to deficient seeding of the bone marrow. To address this possibility, we examined the consequences of loss of Fancd2, a central component of the FA pathway. Examination of embryonic day 14.5 (E14.5) Fancd2 knockout (KO) fetal livers showed a decrease in total cellularity and specific declines in long-term and short-term hematopoietic stem cell (LT-HSC and ST-HSC, respectively) numbers. Fancd2 KO fetal liver cells display similar functional defects to Fancd2 adult bone marrow cells, including reduced colony-forming units, increased mitomycin C sensitivity, increased LT-HSC apoptosis, and heavily impaired competitive repopulation, implying that these defects are intrinsic to the fetal liver and are not dependent on the accumulation of DNA damage during aging. Telomere shortening, an aging-related mechanism proposed to contribute to HSC apoptosis and bone marrow failure in FA, was not observed in Fancd2 KO fetal livers. In summary, loss of Fancd2 yields significant defects to fetal liver hematopoiesis, particularly the HSC population, which mimics key phenotypes from adult Fancd2 KO bone marrow independently of aging-accrued DNA damage.
Collapse
|
50
|
Abstract
The recent advent of the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-CRISPR associated protein 9 (Cas9) system for precise genome editing has revolutionized methodologies in haematology and oncology studies. CRISPR-Cas9 technology can be used to remove and correct genes or mutations, and to introduce site-specific therapeutic genes in human cells. Inherited haematological disorders represent ideal targets for CRISPR-Cas9-mediated gene therapy. Correcting disease-causing mutations could alleviate disease-related symptoms in the near future. The CRISPR-Cas9 system is also a useful tool for delineating molecular mechanisms involving haematological malignancies. Prior to the use of CRISPR-Cas9-mediated gene correction in humans, appropriate delivery systems with higher efficiency and specificity must be identified, and ethical guidelines for applying the technology with controllable safety must be established. Here, the latest applications of CRISPR-Cas9 technology in haematological disorders, current challenges and future directions are reviewed and discussed.
Collapse
Affiliation(s)
- Han Zhang
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas-Health Science Centre at Houston, Houston, TX, USA
| | - Nami McCarty
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), University of Texas-Health Science Centre at Houston, Houston, TX, USA.
| |
Collapse
|