1
|
Zahr RS, Kang G, Zhang X, Rashkin SR, Kovesdy CP, Takemoto C, Weiss M, Lebensburger J, Ataga KI, Saraf SL. Development of Polygenic Risk Score for Persistent Albuminuria in Children and Adults With Sickle Cell Anemia. Am J Hematol 2025; 100:1019-1028. [PMID: 40186439 PMCID: PMC12068958 DOI: 10.1002/ajh.27678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 04/07/2025]
Abstract
Albuminuria is associated with high-risk apolipoprotein-L1 variants (APOL1 G1/G2) in patients with sickle cell anemia (SCA). However, this gene variant does not account for all chronic kidney disease (CKD) risk. We hypothesized that we could develop a polygenic risk score (PRS) for CKD in SCA, combining APOL1 G1/G2 with other candidate genes that modify SCA severity and further stratify patients into risk categories based on this risk score. Variants in APOL1, HMOX1 (rs743811), BCL11A (rs1424407), and α-thalassemia (α -3.7) were identified in children with SCA enrolled in the Sickle Cell Clinical Research and Intervention Program longitudinal cohort (SCCRIP). We individually tested the association of these variants with persistent albuminuria, tested a three-variant PRS (PRS-3) (APOL1, BCL11A (rs1424407), and α-3.7), and developed a four-variant PRS (PRS-4) after adding HMOX1 (rs743811) to PRS-3 using the summation of high-risk alleles. An adult SCA cohort from the University of Illinois, Chicago (UIC), was used for validation. Persistent albuminuria was defined as having a urine albumin-to-creatinine ratio (UACR) ≥ 30 mg/g on at least 2 of 3 consecutive measurements. In both cohorts, APOL1 risk variants increased the risk while α-thalassemia protected against persistent albuminuria. PRS-4 was significantly associated with persistent albuminuria (SCCRIP: p = 0.004; UIC: p = 0.00016). When stratifying patients into three and four risk categories based on the PRS, 58% and 86% of the high-risk (PRS-3) and 54% and 89% of very high-risk (PRS-4) categories developed persistent albuminuria cases in the SCCRIP and UIC cohorts, respectively. A PRS may identify high-risk SCA patients for albuminuria. Applying this PRS to guide the early implementation of disease modifiers and renoprotective therapies may help reduce the burden of SCA-related CKD. Trial Registration: NCT02098863.
Collapse
Affiliation(s)
- Rima S. Zahr
- Department of Paediatric Nephrology and Hypertension, UTHSC, Memphis, TN
| | - Guolian Kang
- Department of Biostatistics, St. Jude Children’s Research Hospital Memphis, TN
| | - Xu Zhang
- Department of Haematology and Oncology, Sickle Cell Center, UIC, Chicago, IL
| | - Sara R. Rashkin
- Department of Hematology, St. Jude Children’s Research Hospital Memphis, TN
| | | | - Clifford Takemoto
- Department of Hematology, St. Jude Children’s Research Hospital Memphis, TN
| | - Mitch Weiss
- Department of Hematology, St. Jude Children’s Research Hospital Memphis, TN
| | | | | | - Santosh L. Saraf
- Department of Haematology and Oncology, Sickle Cell Center, UIC, Chicago, IL
| |
Collapse
|
2
|
Butt H, Mandava M, Jacobsohn D. Advances in Gene Therapy for Sickle Cell Disease: From Preclinical Innovations to Clinical Implementation and Access Challenges. CRISPR J 2025. [PMID: 40356202 DOI: 10.1089/crispr.2024.0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025] Open
Abstract
Sickle cell disease (SCD) is a hereditary blood disorder caused by a specific mutation in the β-globin gene, leading to the production of hemoglobin S, which deforms red blood cells, causing occlusion in small blood vessels. This results in pain, anemia, organ damage, infections, and increased stroke risk. Treatment options, including disease-modifying therapies and curative hematopoietic stem cell transplants, have limited accessibility. Recently, autologous gene therapy has emerged as a promising curative option, particularly for SCD. Gene editing techniques such as CRISPR, base editing, and prime editing offer potential to correct this mutation. In this review, we discuss recent preclinical studies and clinical trials of gene and cell therapies, focusing on the progress of FDA-approved treatments like Lyfgenia and Casgevy. We also examine the many challenges, including accessibility, safety, and long-term efficacy, which continue to shape the future of SCD gene therapy.
Collapse
Affiliation(s)
- Henna Butt
- Cancer and Blood Disorders Center, Children's National Hospital, Washington, District of Columbia, USA
- George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Mamatha Mandava
- Cancer and Blood Disorders Center, Children's National Hospital, Washington, District of Columbia, USA
- George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - David Jacobsohn
- Cancer and Blood Disorders Center, Children's National Hospital, Washington, District of Columbia, USA
- George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| |
Collapse
|
3
|
Ball J, Bradley A, Le A, Tisdale JF, Uchida N. Current and future treatments for sickle cell disease: From hematopoietic stem cell transplantation to in vivo gene therapy. Mol Ther 2025; 33:2172-2191. [PMID: 40083162 DOI: 10.1016/j.ymthe.2025.03.016] [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: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
Sickle cell disease (SCD) is a single-gene disorder caused by a point mutation of the β-globin gene, resulting in hemolytic anemia, acute pain, multiorgan damage, and early mortality. Hydroxyurea is a first-line drug therapy that switches sickle-globin to non-pathogenic γ-globin; however, it requires lifelong oral administration. Allogeneic hematopoietic stem cell (HSC) transplantation allows for a one-time cure for SCD, albeit with histocompatibility limitations. Therefore, autologous HSC gene therapy was developed to cure SCD in a single treatment, without HSC donors. Current HSC gene therapy is based on the ex vivo culture of patients' HSCs with lentiviral gene addition and gene editing, followed by autologous transplantation back to the patient. However, the complexity of the treatment process and high costs hinder the universal application of ex vivo gene therapy. Therefore, the development of in vivo HSC gene therapy, where gene therapy tools are directly administered to patients, is desirable to provide a more accessible, cost-effective solution that can cure SCD worldwide. In this review, we discuss current treatments, including drug therapies, HSC transplantation, and ex vivo gene therapy; the development of gene therapy tools; and progress toward curative in vivo gene therapy in SCD.
Collapse
Affiliation(s)
- Julia Ball
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Avery Bradley
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Anh Le
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
4
|
Williams DA, Kohn DB, Thrasher AJ. Ex vivo modification of hematopoietic stem and progenitor cells for gene therapy. Mol Ther 2025; 33:2141-2153. [PMID: 40176348 DOI: 10.1016/j.ymthe.2025.03.058] [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: 02/20/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025] Open
Abstract
The development of viral vectors has been particularly critical for genetic therapies of hematological diseases. Before the development of retrovirus vectors (RVVs), gene transfer into mammalian cells was accomplished by transduction of DNA plasmids by chemical means and later by electroporation. The main limitation of these methods is the inefficiency of transfer of intact sequences, and particularly with electroporation significant cell death of the manipulated cells. The earliest successful human gene therapy trials utilized γ-RVVs and many of the techniques developed in the 1980s. A breakthrough for the field was the exploitation and development of HIV for transfer vectors, termed lentivirus vectors. In this review, we highlight uses of retro- and lentivirus vectors in monogenic diseases in which hematopoietic stem cells are used in the autologous setting to treat immunodeficiencies, hemoglobinopathies and metabolic diseases. The three authors' perspective represent experiences in the field over four decades that encompasses both basic translational research and development and oversight of early and ongoing gene therapy trials utilizing viral vectors.
Collapse
Affiliation(s)
- David A Williams
- Boston Children's Hospital, Dana-Farber & Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA 02115 USA.
| | - Donald B Kohn
- Department of Pediatrics (Hematology/Oncology), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Adrian J Thrasher
- Molecular and Cellular Immunology, GOS Institute of Child Health, University College London, London WC1N 1EH, UK
| |
Collapse
|
5
|
Kerner G, Kamitaki N, Strober B, Price AL. Mapping disease loci to biological processes via joint pleiotropic and epigenomic partitioning. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.05.25327017. [PMID: 40385425 PMCID: PMC12083580 DOI: 10.1101/2025.05.05.25327017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Genome-wide association studies (GWAS) have identified thousands of disease-associated loci, yet their interpretation remains limited by the heterogeneity of underlying biological processes. We propose Joint Pleiotropic and Epigenomic Partitioning (J-PEP), a clustering framework that integrates pleiotropic SNP effects on auxiliary traits and tissue-specific epigenomic data to partition disease-associated loci into biologically distinct clusters. To benchmark J-PEP against existing methods, we introduce a metric-Pleiotropic and Epigenomic Prediction Accuracy (PEPA)-that evaluates how well the clusters predict SNP-to-trait and SNP-to-tissue associations using off-chromosome data, avoiding overfitting. Applying J-PEP to GWAS summary statistics for 165 diseases/traits (average N = 290 K ), we attained 16-30% higher PEPA than pleiotropic or epigenomic partitioning approaches with larger improvements for well-powered traits, consistent with simulations; these gains arise from J-PEP's tendency to upweight correlated structure-signals present in both auxiliary trait and tissue data-thereby emphasizing shared components. For type 2 diabetes (T2D), J-PEP identified clusters refining canonical pathological processes while revealing underexplored immune and developmental signals. For hypertension (HTN), J-PEP identified stromal and adrenal-endocrine processes that were not identified in prior analyses. For neutrophil count, J-PEP identified hematopoietic, hepatic-inflammatory, and neuroimmune processes, expanding biological interpretation beyond classical immune regulation. Notably, integrating single-cell chromatin accessibility data refined bulk-based clusters, enhancing cell-type resolution and specificity. For T2D, single-cell data refined a bulk endocrine cluster to pancreatic islet β -cells, consistent with established β -cell dysfunction in insulin deficiency; for HTN, single-cell data refined a bulk endocrine cluster to adrenal cortex cells, consistent with a GO enrichment for neutrophil-mediated inflammation that implicates feedback between aldosterone production in the adrenal gland and local immune signaling. In conclusion, J-PEP provides a principled framework for partitioning GWAS loci into interpretable, tissue-informed clusters that provide biological insights on complex disease.
Collapse
Affiliation(s)
- Gaspard Kerner
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Nolan Kamitaki
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
| | - Benjamin Strober
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Alkes L Price
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Broad Institute of MIT and Harvard, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
6
|
Fu L, Upadhyay R, Pokrovskii M, Chen FM, Romero-Meza G, Griesemer A, Littman DR. PRDM16-dependent antigen-presenting cells induce tolerance to gut antigens. Nature 2025:10.1038/s41586-025-08982-4. [PMID: 40228524 DOI: 10.1038/s41586-025-08982-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
The gastrointestinal tract is continuously exposed to foreign antigens in food and commensal microorganisms with potential to induce adaptive immune responses. Peripherally induced T regulatory (pTreg) cells are essential for mitigating inflammatory responses to these agents1-4. Although RORγt+ antigen-presenting cells (APCs) have been shown to programme gut microbiota-specific pTreg cells5-7, their definition remains incomplete, and the APC responsible for food tolerance has remained unknown. Here we identify an APC subset that is required for differentiation of both food- and microbiota-specific pTreg cells and for establishment of oral tolerance. Development and function of these APCs require expression of the transcription factors PRDM16 and RORγt, as well as a unique Rorc(t) cis-regulatory element. Gene expression, chromatin accessibility, and surface marker analysis establish the pTreg-inducing APCs as myeloid in origin, distinct from type 3 innate lymphoid cells, and sharing epigenetic profiles with classical dendritic cells, and designate them PRDM16+RORγt+ tolerizing dendritic cells (tolDCs). Upon genetic perturbation of tolDCs, we observe a substantial increase in food antigen-specific T helper 2 cells in lieu of pTreg cells, leading to compromised tolerance in mouse models of asthma and food allergy. Single-cell analyses of freshly resected mesenteric lymph nodes from a human organ donor, as well as multiple specimens of human intestine and tonsil, reveal candidate tolDCs with co-expression of PRDM16 and RORC and an extensive transcriptome shared with tolDCs from mice, highlighting an evolutionarily conserved role across species. Our findings suggest that a better understanding of how tolDCs develop and how they regulate T cell responses to food and microbial antigens could offer new insights into developing therapeutic strategies for autoimmune and allergic diseases as well as organ transplant tolerance.
Collapse
Affiliation(s)
- Liuhui Fu
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Rabi Upadhyay
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Maria Pokrovskii
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Calico Life Sciences, South San Francisco, CA, USA
| | - Francis M Chen
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Gabriela Romero-Meza
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| | - Adam Griesemer
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Dan R Littman
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA.
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| |
Collapse
|
7
|
Ilboudo Y, Brosseau N, Lo KS, Belhaj H, Moutereau S, Marshall K, Reid M, Kutlar A, Ashley-Koch AE, Telen MJ, Joly P, Galactéros F, Bartolucci P, Lettre G. A replication study of novel fetal hemoglobin-associated genetic variants in sickle cell disease-only cohorts. Hum Mol Genet 2025; 34:699-710. [PMID: 39886999 PMCID: PMC11973897 DOI: 10.1093/hmg/ddaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/18/2024] [Accepted: 01/23/2024] [Indexed: 02/01/2025] Open
Abstract
Sickle cell disease (SCD) is the most common monogenic disease in the world and is caused by mutations in the β-globin gene (HBB). Notably, SCD is characterized by extreme clinical heterogeneity. Inter-individual variation in fetal hemoglobin (HbF) levels strongly contributes to this patient-to-patient variability, with high HbF levels associated with decreased morbidity and mortality. Genetic association studies have identified and replicated HbF levels-associated variants at three loci: BCL11A, HBS1L-MYB, and HBB. In SCD patients, genetic variation at these three loci accounts for ~ 50% of HbF heritability. Genome-wide association studies (GWAS) in non-anemic and SCD patients of multiple ancestries have identified 20 new HbF-associated variants. However, these genetic associations have yet to be replicated in independent SCD cohorts. Here, we validated the association between HbF levels and variants at five of these new loci (ASB3, BACH2, PFAS, ZBTB7A, and KLF1) in up to 3740 SCD patients. By combining CRISPR inhibition and single-cell transcriptomics, we also showed that sequences near non-coding genetic variants at BACH2 (rs4707609) and KLF1 (rs2242514, rs10404876) can control the production of the β-globin genes in erythroid HUDEP-2 cells. Finally, we analyzed whole-exome sequence data from 1354 SCD patients but could not identify rare genetic variants of large effect on HbF levels. Together, our results confirm five new HbF-associated loci that can be functionally studied to develop new strategies to induce HbF expression in SCD patients.
Collapse
Affiliation(s)
- Yann Ilboudo
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
- Department of Medicine, Université de Montréal, 2900 Boul. Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Nicolas Brosseau
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
- Department of Medicine, Université de Montréal, 2900 Boul. Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Ken Sin Lo
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
- Department of Medicine, Université de Montréal, 2900 Boul. Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Hicham Belhaj
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
- Department of Medicine, Université de Montréal, 2900 Boul. Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Stéphane Moutereau
- Red Blood Cell Laboratory, Department of Biochemistry-Pharmacology, Hôpital Universitaire Henri Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Est, IMRB - U955 - Équipe no 2, Créteil, France
| | - Kwesi Marshall
- Tropical Metabolism Research Unit (TMRU), Caribbean Institute for Health Research (CAIHR), University of the West Indies, Mona, Kingston 7, Jamaica
| | - Marvin Reid
- Graduate Studies and Research, University of the West Indies, Mona, Kingston 7, Jamaica
| | - Abdullah Kutlar
- Center for Blood Disorders, Augusta University, Augusta, Georgia 30912, USA
| | - Allison E Ashley-Koch
- Department of Medicine, Duke University Medical Center, Durham, NC 27707, USA
- Duke Molecular Physiology Institute, Duke University Medical Center, 300 North Duke Street, Durham, NC 27701, USA
| | - Marilyn J Telen
- Duke Comprehensive Sickle Cell Center and Division of Hematology, Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Philippe Joly
- Unité Fonctionnelle 34445 ‘Biochimie des Pathologies Érythrocytaires’, Laboratoire de Biochimie et Biologie Moléculaire Grand-Est, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
- Laboratoire Inter-Universitaire de Biologie de la Motricité (LIBM) EA7424, Equipe ‘Biologie Vasculaire et du Globule Rouge’, Université Claude Bernard Lyon 1, Comité d’Universités et d’Établissements (COMUE), Lyon, France
| | - Frédéric Galactéros
- Red Cell Genetic Disease Unit, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Est, IMRB - U955 - Équipe no 2, Créteil, France
| | - Pablo Bartolucci
- Red Cell Genetic Disease Unit, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Est, IMRB - U955 - Équipe no 2, Créteil, France
| | - Guillaume Lettre
- Montreal Heart Institute, 5000 Bélanger Street, Montréal, Québec, H1T 1C8, Canada
- Department of Medicine, Université de Montréal, 2900 Boul. Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| |
Collapse
|
8
|
Martin-Rufino JD, Caulier A, Lee S, Castano N, King E, Joubran S, Jones M, Goldman SR, Arora UP, Wahlster L, Lander ES, Sankaran VG. Transcription factor networks disproportionately enrich for heritability of blood cell phenotypes. Science 2025; 388:52-59. [PMID: 40179192 DOI: 10.1126/science.ads7951] [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: 08/28/2024] [Accepted: 02/12/2025] [Indexed: 04/05/2025]
Abstract
Most phenotype-associated genetic variants map to noncoding regulatory regions of the human genome, but their mechanisms remain elusive in most cases. We developed a highly efficient strategy, Perturb-multiome, to simultaneously profile chromatin accessibility and gene expression in single cells with CRISPR-mediated perturbation of master transcription factors (TFs). We examined the connection between TFs, accessible regions, and gene expression across the genome throughout hematopoietic differentiation. We discovered that variants within TF-sensitive accessible chromatin regions in erythroid differentiation, although representing <0.3% of the genome, show a ~100-fold enrichment for blood cell phenotype heritability, which is substantially higher than that for other accessible chromatin regions. Our approach facilitates large-scale mechanistic understanding of phenotype-associated genetic variants by connecting key cis-regulatory elements and their target genes within gene regulatory networks.
Collapse
Affiliation(s)
- Jorge Diego Martin-Rufino
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Alexis Caulier
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Seayoung Lee
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Nicole Castano
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Emily King
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Samantha Joubran
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Marcus Jones
- Nascent Transcriptomics Core, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Seth R Goldman
- Nascent Transcriptomics Core, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Uma P Arora
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Lara Wahlster
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Eric S Lander
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| |
Collapse
|
9
|
Long E, Williams J, Zhang H, Choi J. An evolving understanding of multiple causal variants underlying genetic association signals. Am J Hum Genet 2025; 112:741-750. [PMID: 39965570 DOI: 10.1016/j.ajhg.2025.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/20/2025] Open
Abstract
Understanding how genetic variation contributes to phenotypic variation is a fundamental question in genetics. Genome-wide association studies (GWASs) have discovered numerous genetic associations with various human phenotypes, most of which contain co-inherited variants in strong linkage disequilibrium (LD) with indistinguishable statistical significance. The experimental and analytical difficulty in identifying the "causal variant" among the co-inherited variants has traditionally led mechanistic studies to focus on relatively simple loci, where a single functional variant is presumed to explain most of the association signal and affect a target gene. The notion that a single causal variant is responsible for an association signal, while other variants in LD are merely correlated, has often been assumed in functional studies. However, emerging evidence powered by high-throughput experimental tools and context-specific functional databases argues that even a single independent signal may involve multiple functional variants in strong LD, each contributing to the observed genetic association. In this perspective, we articulate this evolving understanding of causal variants through examples from both traditional locus-by-locus approaches and more recent high-throughput functional studies. We then discuss the implications and prospects of this notion in understanding the genetic architecture of complex traits and interpreting the variant-level causality in GWAS follow-up studies.
Collapse
Affiliation(s)
- Erping Long
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jacob Williams
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Haoyu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
10
|
Wallace KA, Gerstenberg TL, Ennis CL, Perez-Bermejo JA, Partridge JR, Bandoro C, Matern WM, Andreoletti G, Krassovsky K, Kabir S, Lalisan CD, Churi AR, Chew GM, Corbo L, Vincelette JE, Klasson TD, Silva BJ, Strukov YG, Quejarro BJ, Hill KA, Treusch S, Grogan JL, Dever DP, Porteus MH, Wienert B. A differentiated β-globin gene replacement strategy uses heterologous introns to restore physiological expression. Mol Ther 2025; 33:1407-1419. [PMID: 40022449 PMCID: PMC11997512 DOI: 10.1016/j.ymthe.2025.02.036] [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: 07/03/2024] [Revised: 11/19/2024] [Accepted: 02/25/2025] [Indexed: 03/03/2025] Open
Abstract
β-Hemoglobinopathies are common monogenic disorders. In sickle cell disease (SCD), a single mutation in the β-globin (HBB) gene results in dysfunctional hemoglobin protein, while in β-thalassemia, over 300 mutations distributed across the gene reduce β-globin levels and cause severe anemia. Genetic engineering replacing the whole HBB gene through homology-directed repair (HDR) is an ideal strategy to restore a benign genotype and rescue HBB expression for most genotypes. However, this is technically challenging because (1) the insert must not be homologous to the endogenous gene and (2) synonymous codon-optimized, intron-less sequences may not reconstitute adequate β-globin levels. Here, we developed an HBB gene replacement strategy using CRISPR-Cas9 that successfully addresses these challenges. We determined that a DNA donor containing a diverged HBB coding sequence and heterologous introns to avoid sequence homology provides proper physiological expression. We identified a DNA donor that uses truncated γ-globin introns, results in 34% HDR, and rescues β-globin expression in in vitro models of SCD and β-thalassemia in hematopoietic stem and progenitor cells (HSPCs). Furthermore, while HDR allele frequency dropped in vivo, it was maintained at ∼15%, demonstrating editing of long-term repopulating HSPCs. In summary, our HBB gene replacement strategy offers a differentiated approach by restoring naturally regulated adult hemoglobin expression.
Collapse
Affiliation(s)
- Kirby A Wallace
- Graphite Bio, Inc., South San Francisco, CA 94080, USA; Kamau Therapeutics, Inc., South San Francisco, CA 94080, USA
| | | | - Craig L Ennis
- Graphite Bio, Inc., South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | - Shaheen Kabir
- Graphite Bio, Inc., South San Francisco, CA 94080, USA
| | | | | | - Glen M Chew
- Graphite Bio, Inc., South San Francisco, CA 94080, USA
| | - Lana Corbo
- Graphite Bio, Inc., South San Francisco, CA 94080, USA
| | | | | | - Brian J Silva
- Graphite Bio, Inc., South San Francisco, CA 94080, USA
| | | | | | - Kaisle A Hill
- Graphite Bio, Inc., South San Francisco, CA 94080, USA
| | | | - Jane L Grogan
- Graphite Bio, Inc., South San Francisco, CA 94080, USA
| | | | | | - Beeke Wienert
- Graphite Bio, Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
11
|
Chen L, Liu D, Hong W, Xu L, Cheng L, Luo Y, Xu H, Liang J, Fang J, Li X. Creating New Cis-Regulatory Elements of HBD to Reactivate Delta-Globin. Hum Gene Ther 2025; 36:765-773. [PMID: 40114594 DOI: 10.1089/hum.2024.186] [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] [Indexed: 03/22/2025] Open
Abstract
β-thalassemia and sickle cell disease (SCD) are global monogenic blood system disorders, and reactivated δ-globin is expected to replace missing or abnormal β-globin. With the development of gene editing technology, activating γ-globin for treating β-thalassemia and SCD has been highly successful. However, δ-globin, as another important potential therapeutic target, has few related studies. Gene editing technology introduced cis-acting elements, including NF-Y, KLF1, GATA1, and TAL1, into the regulatory region of HBD, successfully activating the expression of δ-globin. It was confirmed that the activation effect of δ-globin was closely related to the location of the introduced cis-acting elements. In this study, the mutation creates a de novo binding site for KLF1 at -85∼93 bp upstream of the transcription start site of the HBD gene, as well as the site for TAL1 and GATA1 cobinding motifs at -59 to ∼-78 bp, which could effectively activate δ-globin.
Collapse
Affiliation(s)
- Lini Chen
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Diandian Liu
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Weicong Hong
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Luhong Xu
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Lin Cheng
- Reforgene Medicine, Guangzhou, People's Republic of China
| | - Ying Luo
- Reforgene Medicine, Guangzhou, People's Republic of China
| | - Hui Xu
- Reforgene Medicine, Guangzhou, People's Republic of China
| | - Junbin Liang
- Reforgene Medicine, Guangzhou, People's Republic of China
| | - Jianpei Fang
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xinyu Li
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| |
Collapse
|
12
|
Fu L, Upadhyay R, Pokrovskii M, Chen FM, Romero-Meza G, Griesemer A, Littman DR. Prdm16-dependent antigen-presenting cells induce tolerance to intestinal antigens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.23.604803. [PMID: 39091750 PMCID: PMC11291166 DOI: 10.1101/2024.07.23.604803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The gastrointestinal tract is continuously exposed to foreign antigens in food and commensal microbes with potential to induce adaptive immune responses. Peripherally induced T regulatory (pTreg) cells are essential for mitigating inflammatory responses to these agents1-4. While RORγt+ antigen-presenting cells (RORγt-APCs) were shown to program gut microbiota-specific pTreg5-7, their definition remains incomplete, and the APC responsible for food tolerance has remained elusive. Here, we identify a distinct subset of RORγt-APCs, designated tolerogenic dendritic cells (tDC), required for differentiation of both food- and microbiota-specific pTreg cells and for establishment of oral tolerance. tDC development and function require expression of the transcription factors Prdm16 and RORγt, as well as a unique Rorc(t) cis-regulatory element. Gene expression, chromatin accessibility, and surface marker analysis establish tDC as myeloid in origin, distinct from ILC3, and sharing epigenetic profiles with classical DC. Upon genetic perturbation of tDC, we observe a substantial increase in food antigen-specific T helper 2 (Th2) cells in lieu of pTreg, leading to compromised tolerance in mouse models of asthma and food allergy. Single-cell analyses of freshly resected mesenteric lymph nodes from a human organ donor, as well as multiple specimens of human intestine and tonsil, reveal candidate tDC with co-expression of PRDM16 and RORC and an extensive transcriptome shared with mice, highlighting an evolutionarily conserved role across species. Our findings suggest that a better understanding of how tDC develop and how they regulate T cell responses to food and microbial antigens could offer new insights into developing therapeutic strategies for autoimmune and allergic diseases as well as organ transplant tolerance.
Collapse
Affiliation(s)
- Liuhui Fu
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Rabi Upadhyay
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Maria Pokrovskii
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Calico Life Sciences, LLC, South San Francisco, CA, USA
| | - Francis M. Chen
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Gabriela Romero-Meza
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| | - Adam Griesemer
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Dan R. Littman
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| |
Collapse
|
13
|
Perez AR, Mavrothalassitis O, Chen JS, Hellman J, Gropper MA. CRISPR: fundamental principles and implications for anaesthesia. Br J Anaesth 2025; 134:839-852. [PMID: 39855935 PMCID: PMC11867086 DOI: 10.1016/j.bja.2024.11.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/22/2024] [Accepted: 11/01/2024] [Indexed: 01/27/2025] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-based medical therapies are increasingly gaining regulatory approval worldwide. Consequently, patients receiving CRISPR therapy will come under the care of anaesthesiologists. An understanding of CRISPR, its technological implementations, and the characteristics of patients likely to receive this therapy will be essential to caring for this patient population. However, the role of CRISPR in anaesthesiology extends beyond simply caring for patients with prior CRISPR therapy. CRISPR has multiple direct potential applications in anaesthesia, particularly for managing chronic pain and critical illness. Additionally, given the unique skills anaesthesiologists possess, CRISPR potentially allows new roles for anaesthesiologists in the field of oncology. Consequently, CRISPR technology could enable new domains of anaesthetic practice. This review provides a primer on CRISPR for anaesthesiologists and an overview on how the technology could impact the field.
Collapse
Affiliation(s)
- Alexendar R Perez
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Silico Therapeutics, Inc., San Jose, CA, USA.
| | - Orestes Mavrothalassitis
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | | | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Michael A Gropper
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
14
|
de Franceschi L, Locatelli F, Rees D, Chabannon C, Dalle J, Rivella S, Iolascon A, Lobitz S, Abboud MR, de la Fuente J, Flevari P, Angelucci E, de Montalembert M. Selecting patients with sickle cell disease for gene addition or gene editing-based therapeutic approaches: Report on behalf of a joint EHA Specialized Working Group and EBMT Hemoglobinopathies Working Party consensus conference. Hemasphere 2025; 9:e70089. [PMID: 40084235 PMCID: PMC11904809 DOI: 10.1002/hem3.70089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/05/2024] [Accepted: 12/09/2024] [Indexed: 03/16/2025] Open
Abstract
Sickle cell disease (SCD) remains associated with reduced life expectancy and poor quality of life despite improvements observed in the last decades mostly related to comprehensive care, use of hydroxycarbamide, screening to identify patients at risk of strokes, and implementation of safe transfusion protocols. The course of the disease is highly variable, making it difficult to predict severity and response to therapy. Allogeneic hematopoietic stem cell transplantation potentially provides a cure with a relatively low rate of complications, but few patients have an HLA-identical sibling. The hopes of patients and healthcare providers have been raised after the initial excellent results of gene therapy studies. However, there is a strong contrast between the high expectations of families and patients and the limited availability of the product, which is technically complex and very expensive. In light of this consideration and of the limited data available on the long-term efficacy and toxicity of different gene therapy approaches, the European Hematology Association Red Cell & Iron Specialized Working Group (EHA SWG) and the hemoglobinopathy working part of the European Blood & Marrow Transplant (EBMT) Group have prioritized the development of recommendations for selection of patients with SCD who are good candidates for gene therapy. The decision-making algorithm was developed by a panel of experts in hemoglobinopathies and/or transplantation chosen by EHA SWG and EBMT, to discuss the selection of SCD patients for gene therapy and draw notes on the related clinical problems.
Collapse
Affiliation(s)
- Lucia de Franceschi
- Department of Engineering for Innovative MedicineUniversity of VeronaVeronaItaly
- Azienda Ospedaliera Universitaria integrata di VeronaVeronaItaly
| | - Franco Locatelli
- IRCCS Bambino Gesù Children's HospitalCatholic University of the Sacred HeartRomeItaly
| | - David Rees
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences Medicine, King's College London, and Department of Haematological MedicineKing's College HospitalLondonUK
| | - Christian Chabannon
- Institut Paoli‐Calmettes Comprehensive Cancer Center and Module Biotherapies du Centre d'Investigations Cliniques de Marseille, INSERM‐Aix‐Marseille Université AP‐HM‐IPCCBT‐1409MarseilleFrance
| | - Jean‐Hugues Dalle
- Pediatric Hematology and Immunoloy Department, Robert‐Debré Academic HospitalGHU AP‐HP Nord Université Paris CitéParisFrance
| | - Stefano Rivella
- Department of PediatricsHematology, The Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
- Penn Institute for RNA InnovationUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Penn Institute for Regenerative Medicine (IRM)University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli Federico IINaplesItaly
- CEINGE Biotecnologie AvanzateNaplesItaly
| | - Stephan Lobitz
- Pediatric Hematology & Oncology, Gemeinschaftsklinikum MittelrheinKoblenzGermany
| | - Miguel R. Abboud
- Department of Pediatrics and Adolescent MedicineAmerican University of BeirutBeirutLebanon
| | - Josu de la Fuente
- Department of Immunology and InflammationCentre for Haematology, Imperial College LondonLondonUK
- Department of PaediatricsImperial College Healthcare NHS TrustLondonUK
| | - Pagona Flevari
- Thalassemia Unit—Center of Expertise in Haemoglobinopathies, Laiko General HospitalAthensGreece
| | - Emanuele Angelucci
- UO Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San MartinoGenovaItaly
| | - Mariane de Montalembert
- Department of General Pediatrics and Pediatric Infectious Diseases, Sickle Cell Center, Necker‐Enfants Malades Hospital, Assistance Publique—Hôpitaux de Paris (AP‐HP)Université Paris CitéParisFrance
- Laboratory of Excellence GR‐ExParisFrance
| |
Collapse
|
15
|
Janoudi T, Jagdale M, Wu M, Gorlla S, Zhang P, Shao Y, Li L, Bowley SR, Marco E, Chang KH. Nonclinical evaluation of HBG1/2 and BCL11A as genome-editing targets for the treatment of β-hemoglobinopathies. Blood Adv 2025; 9:808-813. [PMID: 39637306 PMCID: PMC11869951 DOI: 10.1182/bloodadvances.2024014040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Affiliation(s)
| | | | - Meng Wu
- Editas Medicine Inc, Cambridge, MA
| | | | | | | | - Li Li
- Editas Medicine Inc, Cambridge, MA
| | | | | | | |
Collapse
|
16
|
Habara A. Exploratory Review and In Silico Insights into circRNA and RNA-Binding Protein Roles in γ-Globin to β-Globin Switching. Cells 2025; 14:312. [PMID: 39996784 PMCID: PMC11854342 DOI: 10.3390/cells14040312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
β-globin gene cluster regulation involves complex mechanisms to ensure proper expression and function in RBCs. During development, switching occurs as γ-globin is replaced by β-globin. Key regulators, like BCL11A and ZBTB7A, repress γ-globin expression to facilitate this transition with other factors, like KLF1, LSD1, and PGC-1α; these regulators ensure an orchestrated transition from γ- to β-globin during development. While these mechanisms have been extensively studied, circRNAs have recently emerged as key contributors to gene regulation, but their role in β-globin gene cluster regulation remains largely unexplored. Although discovered in the 1970s, circRNAs have only recently been recognized for their functional roles, particularly in interactions with RNA-binding proteins. Understanding how circRNAs contribute to switching from γ- to β-globin could lead to new therapeutic strategies for hemoglobinopathies, such as sickle cell disease and β-thalassemia. This review uses the circAtlas 3.0 database to explore circRNA expressions in genes related to switching from γ- to β-globin expression, focusing on blood, bone marrow, liver, and spleen. It emphasizes the exploration of the potential interactions between circRNAs and RNA-binding proteins involved in β-globin gene cluster regulatory mechanisms, further enhancing our understanding of β-globin gene cluster expression.
Collapse
Affiliation(s)
- Alawi Habara
- Department of Biochemistry, College of Medicine, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
17
|
Ropero P, Peral M, Sánchez-Martínez LJ, Rochas S, Gómez-Álvarez M, Nieto JM, González FA, Villegas A, Benavente C. Phenotype of sickle cell disease. Correlation of haplotypes and polymorphisms in cluster β, BCL11A, and HBS1L-MYB. Pilot study. Front Med (Lausanne) 2025; 12:1347026. [PMID: 40012971 PMCID: PMC11864215 DOI: 10.3389/fmed.2025.1347026] [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: 11/30/2023] [Accepted: 01/14/2025] [Indexed: 02/28/2025] Open
Abstract
Objective/Background Sickle cell disease (SCD) is a monogenic disease with a highly variable phenotype depending on the amount of fetal hemoglobin (HbF), the main modulator. Variation of HbF levels among patients is genetically regulated. HbF determines both the phenotype of the disease and the response to treatment with the main drug used, hydroxyurea. The efforts of the researchers have focused on discovering the genetic factors responsible for HbF variation, mainly describing the haplotypes of the β cluster and single nucleotide polymorphisms (SNPs) at three different loci: BCL11A, HBS1L-MYB, and the β-globin cluster. This study aimed to determine the possible correlation between the number of SNPs and haplotypes with higher HbF levels in a cohort of patients with SCD. A positive association could explain why certain haplotypes, such as Senegal or Arab-Indian, show higher HbF levels and less severe disease. Methods To test this hypothesis, the characterization of haplotypes was performed using the PCR-RFLP technique and genotyping of three SNPs representative of the three loci with the greatest association with HbF variation: XmnI (rs7482144), BCL11A (rs4671393), and HBS1L-MYB (rs9376092). Results We found more SNPs in haplotypes related to higher HbF than those with less HbF, although only the SNP XmnI (rs7482144) showed a statistically significant association. Conclusion We found a direct correlation between haplotypes and the number of SNPs. Haplotypes with higher levels of HbF and less severe phenotypes showed a higher number of SNPs. Thus, the Benin and Bantu haplotypes traditionally associated with poor prognosis showed the fewest mutated SNPs.
Collapse
Affiliation(s)
- Paloma Ropero
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, Madrid, Spain
| | - Miriam Peral
- Instituto de Biología y Genética Molecular, Valladolid, Spain
| | - Luis Javier Sánchez-Martínez
- Departamento de Biodiversidad, Ecología y Evolución, Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Sara Rochas
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, Madrid, Spain
| | - Miguel Gómez-Álvarez
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, Madrid, Spain
| | - Jorge M. Nieto
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, Madrid, Spain
| | - Fernando A. González
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, Madrid, Spain
| | - Ana Villegas
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, Madrid, Spain
| | - Celina Benavente
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, Madrid, Spain
| |
Collapse
|
18
|
Demirci S, Zeng J, Palchaudhuri R, Wu C, Abraham DM, Hayal TB, Essawi K, Nguyen MA, Stasula U, Chu R, Leonard A, Porter SN, Khan MBN, Hinojosa G, Uchida N, Hong S, Lazzarotto CR, Neri NR, da Silva LF, Pellin D, Verma A, Lanieri L, Bhat A, Hammond K, Tate T, Maitland SA, Sheikhsaran F, Bonifacino AC, Krouse AE, Linde NS, Engels T, Golomb J, Tsai SQ, Pruett-Miller SM, Scadden DT, Dunbar CE, Wolfe SA, Donahue RE, Olson LM, Bauer DE, Tisdale JF. BCL11A +58/+55 enhancer-editing facilitates HSPC engraftment and HbF induction in rhesus macaques conditioned with a CD45 antibody-drug conjugate. Cell Stem Cell 2025; 32:209-226.e8. [PMID: 39642886 DOI: 10.1016/j.stem.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 08/03/2024] [Accepted: 10/28/2024] [Indexed: 12/09/2024]
Abstract
Editing the +58 region of the BCL11A erythroid enhancer has shown promise in treating β-globin disorders. To address variations in fetal hemoglobin (HbF) response, we investigated editing both +58 and +55 enhancers. Rhesus macaques transplanted with edited hematopoietic stem/progenitor cells (HSPCs) following busulfan conditioning exhibited durable, high-level (∼90%) editing frequencies post transplantation with sustained HbF reactivation over 4 years, without hematological perturbations. HbF levels were further boosted by stress erythropoiesis or hydroxyurea. Bone marrow analysis revealed that gene edits were predominantly programmed deletions, programmed inversions, and short indels, each disrupting the enhancer core TGN7-9WGATAR half E-box/GATA binding motifs. Nonprogrammed long deletions were disfavored in engrafting cells. CD45 antibody-drug conjugate (ADC) conditioning achieved comparable engraftment and HbF reactivation, whereas lentiviral vector tracking showed polyclonal reconstitution with dynamics similar to animals conditioned with total body irradiation (TBI) or busulfan. Joining CD45-ADC conditioning with combined enhancer editing presents an effective strategy for β-hemoglobinopathies, enabling durable HbF reactivation without chemotherapy.
Collapse
Affiliation(s)
- Selami Demirci
- National Heart, Lung, and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| | - Jing Zeng
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Chuanfeng Wu
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20814, USA
| | - Diana M Abraham
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20814, USA
| | - Taha B Hayal
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20814, USA
| | - Khaled Essawi
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan 45142, Saudi Arabia
| | - My Anh Nguyen
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ulana Stasula
- National Heart, Lung, and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Rebecca Chu
- National Heart, Lung, and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Alexis Leonard
- National Heart, Lung, and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Shaina N Porter
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Muhammad Behroz Naeem Khan
- National Heart, Lung, and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Gabriela Hinojosa
- National Heart, Lung, and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Naoya Uchida
- National Heart, Lung, and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Sogun Hong
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20814, USA
| | - Cicera R Lazzarotto
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Nola R Neri
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Lucas Ferreira da Silva
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Danilo Pellin
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Archana Verma
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Anjali Bhat
- Magenta Therapeutics, Cambridge, MA 02139, USA
| | | | | | - Stacy A Maitland
- Department of Molecular, Cell and Cancer Biology, Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Fatemeh Sheikhsaran
- National Heart, Lung, and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | - Aylin C Bonifacino
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20814, USA
| | - Allen E Krouse
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20814, USA
| | - Nathaniel S Linde
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20814, USA
| | - Theresa Engels
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20814, USA
| | - Justin Golomb
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20814, USA
| | - Shengdar Q Tsai
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David T Scadden
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Cynthia E Dunbar
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20814, USA
| | - Scot A Wolfe
- Department of Molecular, Cell and Cancer Biology, Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Robert E Donahue
- National Heart, Lung, and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20814, USA
| | | | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| | - John F Tisdale
- National Heart, Lung, and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| |
Collapse
|
19
|
Zeng J, Nguyen MA, Liu P, da Silva LF, Levesque S, Lin LY, Justus DG, Petri K, Clement K, Porter SN, Verma A, Neri NR, Rosanwo T, Ciuculescu MF, Abriss D, Mintzer E, Maitland SA, Demirci S, Cha HJ, Orkin SH, Tisdale JF, Williams DA, Zhu LJ, Pruett-Miller SM, Pinello L, Joung JK, Pattanayak V, Manis JP, Armant M, Pellin D, Brendel C, Wolfe SA, Bauer DE. Gene editing without ex vivo culture evades genotoxicity in human hematopoietic stem cells. Cell Stem Cell 2025; 32:191-208.e11. [PMID: 39672163 PMCID: PMC11805672 DOI: 10.1016/j.stem.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/05/2024] [Accepted: 11/01/2024] [Indexed: 12/15/2024]
Abstract
Gene editing the BCL11A erythroid enhancer is a validated approach to fetal hemoglobin (HbF) induction for β-hemoglobinopathy therapy, though heterogeneity in edit allele distribution and HbF response may impact its safety and efficacy. Here, we compare combined CRISPR-Cas9 editing of the BCL11A +58 and +55 enhancers with leading gene modification approaches under clinical investigation. Dual targeting of the BCL11A +58 and +55 enhancers with 3xNLS-SpCas9 and two single guide RNAs (sgRNAs) resulted in superior HbF induction, including in sickle cell disease (SCD) patient xenografts, attributable to simultaneous disruption of core half E-box/GATA motifs at both enhancers. Unintended on-target outcomes of double-strand break (DSB) repair in hematopoietic stem and progenitor cells (HSPCs), such as long deletions and centromere-distal chromosome fragment loss, are a byproduct of cellular proliferation stimulated by ex vivo culture. Editing quiescent HSPCs bypasses long deletion and micronuclei formation and preserves efficient on-target editing and engraftment function.
Collapse
Affiliation(s)
- Jing Zeng
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - My Anh Nguyen
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Pengpeng Liu
- Department of Molecular, Cell and Cancer Biology, Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Lucas Ferreira da Silva
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Molecular Pathology Unit, Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02129, USA; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Sébastien Levesque
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Linda Y Lin
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - David G Justus
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Program in Transfusion Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Karl Petri
- Molecular Pathology Unit, Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02129, USA; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Kendell Clement
- Molecular Pathology Unit, Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02129, USA; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Shaina N Porter
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Archana Verma
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Nola R Neri
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Tolulope Rosanwo
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Daniela Abriss
- TransLab, Boston Children's Hospital, Boston, MA 02115, USA
| | - Esther Mintzer
- Department of Molecular, Cell and Cancer Biology, Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Stacy A Maitland
- Department of Molecular, Cell and Cancer Biology, Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Selami Demirci
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Hye Ji Cha
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Stuart H Orkin
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - David A Williams
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, Department of Molecular Medicine, Program in Bioinformatics and Integrative Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Luca Pinello
- Molecular Pathology Unit, Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02129, USA; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - J Keith Joung
- Molecular Pathology Unit, Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02129, USA; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Vikram Pattanayak
- Molecular Pathology Unit, Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02129, USA; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - John P Manis
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Myriam Armant
- TransLab, Boston Children's Hospital, Boston, MA 02115, USA
| | - Danilo Pellin
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Christian Brendel
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Scot A Wolfe
- Department of Molecular, Cell and Cancer Biology, Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Pinglay S, Lalanne JB, Daza RM, Kottapalli S, Quaisar F, Koeppel J, Garge RK, Li X, Lee DS, Shendure J. Multiplex generation and single-cell analysis of structural variants in mammalian genomes. Science 2025; 387:eado5978. [PMID: 39883753 PMCID: PMC11931979 DOI: 10.1126/science.ado5978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 12/03/2024] [Indexed: 02/01/2025]
Abstract
Studying the functional consequences of structural variants (SVs) in mammalian genomes is challenging because (i) SVs arise much less commonly than single-nucleotide variants or small indels and (ii) methods to generate, map, and characterize SVs in model systems are underdeveloped. To address these challenges, we developed Genome-Shuffle-seq, a method that enables the multiplex generation and mapping of thousands of SVs (deletions, inversions, translocations, and extrachromosomal circles) throughout mammalian genomes. We also demonstrate the co-capture of SV identity with single-cell transcriptomes, facilitating the measurement of SV impact on gene expression. We anticipate that Genome-Shuffle-seq will be broadly useful for the systematic exploration of the functional consequences of SVs on gene expression, the chromatin landscape, and three-dimensional nuclear architecture, while also initiating a path toward a minimal mammalian genome.
Collapse
Affiliation(s)
- Sudarshan Pinglay
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | | | - Riza M. Daza
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Sanjay Kottapalli
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Faaiz Quaisar
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Jonas Koeppel
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
- Wellcome Sanger Institute, Hinxton, UK
| | - Riddhiman K. Garge
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Xiaoyi Li
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - David S. Lee
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| |
Collapse
|
21
|
Shu Y, Chen X, Wei Z, Chen C. Dias-Logan syndrome with a de novo p.Leu360Profs*212 heterozygous pathogenic variant of BCL11A in a Chinese patient: A case report. SAGE Open Med Case Rep 2025; 13:2050313X251314069. [PMID: 39835253 PMCID: PMC11744616 DOI: 10.1177/2050313x251314069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
Dias-Logan syndrome, also known as intellectual developmental disorder with persistence of fetal hemoglobin (HbF), or BCL11A-related intellectual developmental disorder, is an extremely rare neurogenetic disorder characterized by intellectual disability (ID), delayed psychomotor development, variable dysmorphic features, and asymptomatic persistence of fetal hemoglobin. The prevalence and incidence of this condition are currently unknown. We report an 8-year-old Han Chinese male patient with Dias-Logan syndrome who carries a de novo heterozygous pathogenic variant, c.1078dupC (p.Leu360Profs*212), in the BCL11A gene, leading to ID and γ-globin suppression, identified through trio-based whole exome sequencing (trio-WES). All his blood parameters were normal except for an elevated HbF level, which was 19.9% of total hemoglobin. Given the negative family history for ID, epilepsy, and alcohol consumption, de novo inheritance was presumed. Consequently, trio-WES analysis (parents and child) was conducted as it can identify potential new causal variants in the offspring. So far, a comprehensive understanding of the phenotypic spectrum of Dias-Logan syndrome and the impact of genotypic variation on disease severity is still lacking. Therefore, our case report enriches the existing literature on the clinical spectrum and genotype-phenotype correlations of BCL11A-related syndrome and provides some helpful information for diagnosis, management, and genetic counseling.
Collapse
Affiliation(s)
- Yizhuo Shu
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaoling Chen
- Department of Biochemistry, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhuoqun Wei
- Department of Acupuncture and Massage, The Third Clinical Medical College-Rehabilitation College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Chunyue Chen
- Department of Reproductive Medicine, Zhejiang Provincial Hospital of Integrated Traditional Chinese and Western Medicine & Hangzhou Red Cross Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
22
|
Wang Y, Armendariz DA, Wang L, Zhao H, Xie S, Hon GC. Enhancer regulatory networks globally connect non-coding breast cancer loci to cancer genes. Genome Biol 2025; 26:10. [PMID: 39825430 PMCID: PMC11740497 DOI: 10.1186/s13059-025-03474-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Genetic studies have associated thousands of enhancers with breast cancer (BC). However, the vast majority have not been functionally characterized. Thus, it remains unclear how BC-associated enhancers contribute to cancer. RESULTS Here, we perform single-cell CRISPRi screens of 3513 regulatory elements associated with breast cancer to measure the impact of these regions on transcriptional phenotypes. Analysis of > 500,000 single-cell transcriptomes in two breast cancer cell lines shows that perturbation of BC-associated enhancers disrupts breast cancer gene programs. We observe BC-associated enhancers that directly or indirectly regulate the expression of cancer genes. We also find one-to-multiple and multiple-to-one network motifs where enhancers indirectly regulate cancer genes. Notably, multiple BC-associated enhancers indirectly regulate TP53. Comparative studies illustrate subtype specific functions between enhancers in ER + and ER - cells. Finally, we develop the pySpade package to facilitate analysis of single-cell enhancer screens. CONCLUSIONS Overall, we demonstrate that enhancers form regulatory networks that link cancer genes in the genome, providing a more comprehensive understanding of the contribution of enhancers to breast cancer development.
Collapse
Affiliation(s)
- Yihan Wang
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Daniel A Armendariz
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lei Wang
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Huan Zhao
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Shiqi Xie
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Present Address: Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Gary C Hon
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Division of Basic Reproductive Biology Research, Department of Obstetrics and Gynecology, Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
23
|
Dong Z, Ye Y, Zhang W, Luo H, Li J, Zhang Q, Zhang X, Guo X, Xu X. MYB represses ζ-globin expression through upregulating ETO2. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 39757769 DOI: 10.3724/abbs.2024239] [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: 01/07/2025] Open
Abstract
Reactivating the embryonic ζ-globin gene represents a potential therapeutic approach to ameliorate the severe clinical phenotype of α-thalassemia and sickle cell disease. The transcription factor MYB has been extensively proven to be a master regulator of the γ-globin gene, but its role in the regulation of ζ-globin remains incompletely understood. Here, we report a mechanistic study on the derepression of ζ-globin both in vivo and in vitro. We show that MYB depletion in mouse models and human hematopoietic stem cells leads to consistent and remarkable reactivation of ζ-globin. Furthermore, multiomics analysis and functional validation of MYB-knockout and wild-type cell lines reveal that ETO2 functions as a novel repressor of ζ-globin through coordination with NuRD nucleosome remodeling and the deacetylation complex to modulate histone deacetylation of ζ-globin. Additionally, we evaluate the clinical significance of these findings by knocking out ETO2 in primary CD34 + cells from nondeletional hemoglobin H patients, which results in a significant increase in ζ-globin expression. The RNA-seq data reveal that key erythroid genes are more co-regulated by Myb and Eto2 than by Myb and Klf1, highlighting a distinctly enhanced erythroid-specific transcriptional impact within the MYB-ETO2 regulatory axis. Compared with ETO2 knockout alone, codepletion of ETO2 and BCL11A did not significantly activate ζ-globin, suggesting that the MYB-ETO2 pathway primarily silences ζ-globin. Our study reveals a linear MYB-ETO2 signaling pathway crucial for ζ-globin repression and offers new targets for treating α-thalassemia and sickle cell disease.
Collapse
Affiliation(s)
- Zejun Dong
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yuhua Ye
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wei Zhang
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hualei Luo
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jialong Li
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qianqian Zhang
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xinhua Zhang
- Department of Hematology, 923rd Hospital of the People's Liberation Army, Nanning 530021, China
| | - Xiang Guo
- Institute of Blood Diseases, Department of Hematology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xiangmin Xu
- Innovation Center for Diagnostics and Treatment of Thalassemia, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
24
|
Zhang H, Zeng J, Zhang F, Liu J, Liang L. Role of B-Cell Lymphoma/Leukemia 11A in Normal and Malignant Hematopoiesis. BIOLOGY 2025; 14:26. [PMID: 39857257 PMCID: PMC11759832 DOI: 10.3390/biology14010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025]
Abstract
B-cell lymphoma/leukemia 11A (BCL11A) is a crucial transcriptional regulator, widely recognized for its role in controlling fetal hemoglobin and its potential as a gene therapy target for inherited hemoglobinopathies. Beyond this, recent studies have also highlighted its key role in the maturation and function of immune cells and erythrocytes, mediated through the regulation of various molecules during hematopoietic development. The dysregulation of BCL11A disrupts downstream molecular pathways, contributing to the development of several hematological malignancies, particularly leukemias. This review provides a comprehensive overview of the role of BCL11A in normal and malignant hematopoiesis, details the hematological disorders associated with its dysregulation and explores the current therapeutic strategies targeting this transcription factor.
Collapse
Affiliation(s)
- Haihang Zhang
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (H.Z.); (F.Z.)
| | - Junhao Zeng
- Xiangya School of Medicine, Central South University, Changsha 410013, China;
| | - Fangling Zhang
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (H.Z.); (F.Z.)
| | - Jing Liu
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (H.Z.); (F.Z.)
| | - Long Liang
- Department of Hematology, the Second Xiangya Hospital, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China; (H.Z.); (F.Z.)
| |
Collapse
|
25
|
Brusson M, Miccio A. [A CRISPR/Cas approach to β-haemoglobinopathies]. Med Sci (Paris) 2025; 41:33-39. [PMID: 39887096 DOI: 10.1051/medsci/2024191] [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] [Indexed: 02/01/2025] Open
Abstract
Beta-haemoglobinopathies are severe genetic anemias caused by mutations that affect adult haemoglobin production. Many therapeutic approaches aim to reactivate the expression of the fetal hemoglobin genes. To this end, the CRISPR/Cas9 system has recently been used to genetically modify patients' hematopoietic stem/progenitor cells ex vivo and reactivate fetal hemoglobin expression in their erythroid progeny. More than 70 patients with severe β-thalassemia and sickle cell disease have been treated with the Casgevy® therapy. Most have achieved a significant improvement of clinical phenotype, with high editing efficiency in hematopoietic cells associated with normal or near normal hemoglobin levels. While the long-term safety and efficacy of this powerful approach still need to be evaluated, new strategies are being developed to further improve therapeutic outcomes, reduce potential genotoxicity and lower the costs of therapy.
Collapse
Affiliation(s)
- Megane Brusson
- Institut Imagine, Inserm UMR1163, université Paris Cité, Paris, France
| | - Annarita Miccio
- Institut Imagine, Inserm UMR1163, université Paris Cité, Paris, France
| |
Collapse
|
26
|
Cavazzana M, Corsia A, Brusson M, Miccio A, Semeraro M. Treating Sickle Cell Disease: Gene Therapy Approaches. Annu Rev Pharmacol Toxicol 2025; 65:397-413. [PMID: 39259977 DOI: 10.1146/annurev-pharmtox-022124-022000] [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] [Indexed: 09/13/2024]
Abstract
Sickle cell disease (SCD) is a hereditary blood disorder characterized by the presence of abnormal hemoglobin molecules and thus distortion (sickling) of the red blood cells. SCD causes chronic pain and organ damage and shortens life expectancy. Gene therapy emerges as a potentially curative approach for people with SCD who lack a matched sibling donor for hematopoietic stem cell transplantation. Here, we review recent progress in gene therapy for SCD and focus on innovative technologies that target the genetic roots of the disease. We also review the challenges associated with gene therapy, including oncogenic risks, and the need for refined delivery methods. Despite these hurdles, the rapidly evolving landscape of gene therapy for SCD raises hope for a paradigm shift in the treatment of this debilitating disease. As research progresses, a deeper understanding of the molecular mechanisms involved and continuous improvements in gene-editing technologies promise to bring gene therapy for SCD closer to mainstream clinical application, offering a transformative, curative option for patients with this genetic disorder.
Collapse
Affiliation(s)
- Marina Cavazzana
- Imagine Institute, Paris, France
- Centre d'Investigation Clinique en Biothérapie, INSERM UMR1163, Paris, France
- Département de Biothérapie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France;
- Université Paris Cité, Paris, France
| | - Alice Corsia
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
- Université Paris Cité, Paris, France
| | - Megane Brusson
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Annarita Miccio
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Michaela Semeraro
- EA 7323, Université Paris Cité, Pharmacologie et Évaluations Thérapeutiques chez l'Enfant et la Femme Enceinte, Paris, France
- Centre d'Investigation Clinique and Unité de Recherche Clinique, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Cité, Paris, France
| |
Collapse
|
27
|
Ballantine J, Tisdale JF. Gene therapy for sickle cell disease: recent advances, clinical trials and future directions. Cytotherapy 2024:S1465-3249(24)00925-3. [PMID: 39729054 DOI: 10.1016/j.jcyt.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/03/2024] [Indexed: 12/28/2024]
Abstract
Sickle cell disease (SCD) is the most common inherited blood disorder worldwide, impacting millions and imposing severe healthcare challenges, particularly in resource-limited regions. Current treatments have variable efficacy and require lifelong adherence. Allogeneic Hematopoietic Stem Cell Transplantation can be curative but comes with significant side effects and limited donor availability limits its widespread applicability. Gene therapy, by addressing the root genetic causes, offers a revolutionary alternative. This article discusses the molecular mechanisms of SCD and β-thalassemia and highlights advancements in gene therapy, such as gene addition via lentiviral vectors and gene editing with CRISPR/Cas9 technology. Clinical trials have brought about significant progress but challenges remain, including leukemogenesis, delivery efficiency and cost. Future efforts must focus on enhancing efficiency, reducing costs, developing nongenotoxic conditioning regimens and methods for in vivo application.
Collapse
Affiliation(s)
- Josiah Ballantine
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA.
| | - John F Tisdale
- Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
28
|
Li Q, Li X, He S, Li J. Hotspots and status of Fetal Alpha-Thalassemia from 2009 to 2023: a bibliometric analysis. Front Pediatr 2024; 12:1467760. [PMID: 39726529 PMCID: PMC11670076 DOI: 10.3389/fped.2024.1467760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024] Open
Abstract
Objective to evaluate the research status and development hotspots of fetal α-thalassemia by quantitatively analyzing the diagnostic status, key areas, related management measures and prospects of the disease by bibliometrics. Methods The global literature on fetal α-thalassemia and severe α-thalassemia from 2009-2023 in the Web of Science Core Collection (WOSCC) was visually analyzed by VOSviewer and CiteSpace. Results (1) The examination of the quantity of publications concerning fetal α-thalassemia indicates a rising tendency prior to 2018, followed by a decrease after 2018. (2)The United States, China, Italy, Thailand have published more papers, and the United States has more collaborating countries such as Italy and China. (3) Chiang Mai University and Harvard University are the top two institutions with the highest contribution. However, Chiang Mai University's H index (12) and citation frequency per article (8.05) are relatively low and the NC (6,342), H index (33) and citations per article (75.42) of Harvard University are higher than those of the other institutions. (4) Tongsong T, Gambari R and Fucharoen S are the top three prolific authors. Fucharoen S emerges as the most frequently cited author with 738 citations, excluding self-citations. (5) HEMOGLOBIN leading with 87 published papers (NC:601,IF: 0.82, H-index: 13), followed by BLOOD(58 papers, Nc: 3755, IF: 25.48, H-index: 40) and BLOOD CELLS MOLECULES AND DISEASES(39 papers, Nc: 729, IF: 2.37, H-index: 16). (6) The most cited article was published in science and the second and third cited articles were featured in the Proceedings of the National Academy of Sciences; the top 3 clusters of co-cited literature are "gene editing", "polymorphisms", "hydroxyurea". (7) Keywords analysis showe that the top two categories of keyword cluster focus on the prenatal diagnosis and the current treatment strategy of the disease, which remain the research hotspots. Conclusions Recent research on this topic has primarily focused on prenatal diagnosis and treatment strategies. A particular area of interest is the ongoing research on gene therapy.The advances in non-invasive diagnosis and therapeutic methods will change the current management approaches for fetal severe α-thalassemia in the future.
Collapse
Affiliation(s)
- Qiuying Li
- Department of Ultrasonography, Maternity and Child Health Care of Guangxi Zhuang Autonomous Region, Nanning, China
- Graduate School, Guangxi University of Chinese Medicine, Nanning, China
| | - Xinyan Li
- Department of Ultrasonography, Maternity and Child Health Care of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Sheng He
- Birth Defects Prevention and Control Institute of Guangxi Zhuang Autonomous Region, Nanning, China
- Maternity and Child Health Care of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jiao Li
- Maternity and Child Health Care of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
29
|
Frati G, Brusson M, Sartre G, Mlayah B, Felix T, Chalumeau A, Antoniou P, Hardouin G, Concordet JP, Romano O, Turchiano G, Miccio A. Safety and efficacy studies of CRISPR-Cas9 treatment of sickle cell disease highlights disease-specific responses. Mol Ther 2024; 32:4337-4352. [PMID: 39044427 PMCID: PMC11638826 DOI: 10.1016/j.ymthe.2024.07.015] [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: 01/26/2024] [Revised: 05/15/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024] Open
Abstract
Fetal hemoglobin (HbF) reactivation expression through CRISPR-Cas9 is a promising strategy for the treatment of sickle cell disease (SCD). Here, we describe a genome editing strategy leading to reactivation of HbF expression by targeting the binding sites (BSs) for the lymphoma-related factor (LRF) repressor in the γ-globin promoters. CRISPR-Cas9 treatment in healthy donor (HD) and patient-derived HSPCs resulted in a high frequency of LRF BS disruption and potent HbF synthesis in their erythroid progeny. LRF BS disruption did not impair HSPC engraftment and differentiation but was more efficient in SCD than in HD cells. However, SCD HSPCs showed a reduced engraftment and a myeloid bias compared with HD cells. We detected off-target activity and chromosomal rearrangements, particularly in SCD samples (likely because of the higher overall editing efficiency) but did not impact the target gene expression and HSPC engraftment and differentiation. Transcriptomic analyses showed that the editing procedure results in the up-regulation of genes involved in DNA damage and inflammatory responses, which was more evident in SCD HSPCs. This study provides evidence of efficacy and safety for an editing strategy based on HbF reactivation and highlights the need of performing safety studies in clinically relevant conditions, i.e., in patient-derived HSPCs.
Collapse
Affiliation(s)
- Giacomo Frati
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Megane Brusson
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Gilles Sartre
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Bochra Mlayah
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Tristan Felix
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Anne Chalumeau
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Panagiotis Antoniou
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Giulia Hardouin
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Jean-Paul Concordet
- INSERM U1154, CNRS UMR7196, Museum National d'Histoire Naturelle, Paris, France
| | - Oriana Romano
- University of Padova, Department of Molecular Medicine, Padova, Italy
| | | | - Annarita Miccio
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France.
| |
Collapse
|
30
|
Lee H, Friedman MJ, Kim SB, Oh S. DNA regulatory element cooperation and competition in transcription. BMB Rep 2024; 57:509-520. [PMID: 39523506 PMCID: PMC11693600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 11/16/2024] Open
Abstract
Regulation of eukaryotic transcription is a complex process that enables precise temporal and spatial control of gene expression. Promoters, which are cis-regulatory elements (CREs) located proximal to the transcription start site (TSS), selectively integrate regulatory cues from distal CREs, or enhancers, and their associated transcriptional machinery. In this review, we discuss current knowledge regarding CRE cooperation and competition impacting gene expression, including features of enhancer-promoter, enhancer-enhancer, and promoter-promoter interplay. We also provide an overview of recent insights into the underlying molecular mechanisms that facilitate physical and functional interaction of regulatory elements, such as the involvement of enhancer RNAs and biomolecular condensates. [BMB Reports 2024; 57(12): 509-520].
Collapse
Affiliation(s)
- Haram Lee
- Department of Pharmacy, College of Pharmacy, Korea University, Sejong 30019, Korea, Seoul 01795, Korea
| | - Meyer Joseph Friedman
- Department and School of Medicine, University of California, San Diego, CA 92093, USA, Seoul 01795, Korea
| | - Sang Bum Kim
- Department of Pharmacy, College of Pharmacy, Sahmyook University, Seoul 01795, Korea
| | - Soohwan Oh
- Department of Pharmacy, College of Pharmacy, Korea University, Sejong 30019, Korea, Seoul 01795, Korea
| |
Collapse
|
31
|
Wang L, He H, Li Y, Wang X, Yu J, Huang Y, Yu K, He J, Zhao M, Xie T, Li D. BCL11A expression worsens the prognosis of DLBCL and its co-expression with C-MYC predicts poor survival. Pathol Res Pract 2024; 264:155717. [PMID: 39546996 DOI: 10.1016/j.prp.2024.155717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 10/20/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Non-Hodgkin's lymphoma (NHL) is a significant global malignancy, with diffuse large B cell lymphoma (DLBCL) being the most prevalent subtype, accounting for 25-50 % of newly diagnosed cases in China. Despite a 60 % survival rate achieved with R-CHOP regiment for DLBCL, approximately 40 % of patients experience relapse or develop resistance to treatment. While the oncogenic transcription factor B-cell chronic lymphocytic leukaemia/lymphoma 11 A (BCL11A) has been implicated in various tumors, its specific role in DLBCL remains unclear. In this study, we conducted retrospective histomorphological and immunophenotypic analyses on paraffin sample tissues and collected fresh tissue samples for protein and mRNA analyses to investigate the relationship between BCL11A and DLBCL. Additionally, we classified DLBCL into subtypes based on cells of origin (COO) and examined the expressions of BCL11A, C-MYC, P53 and other protein expressions to better understand the factors contributing to poor clinical outcomes in DLBCL. Our findings revealed elevated BCL11A expression in DLBCL, with increased expression associated with worse prognosis and higher C-MYC expression. Patients exhibiting co-expression of C-MYC and BCL11A had significantly lower survival rates compared to those with singular expression. Furthermore, BCL11A protein expression levels demonstrated significant associations with P53 and C-MYC protein expression levels in the Germinal Center B-cell-like (GCB) subtype. These findings suggest that BCL11A may serve as a potential prognostic marker and therapeutic target for DLBCL.
Collapse
Affiliation(s)
- Lixin Wang
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China; Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong He
- Department of Internal Medicine, the First Branch, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanxin Li
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China; Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xingyu Wang
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China; Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jieyang Yu
- Laboratory of Neuropsycholinguistics, Chongqing Medical University, Chongqing, China
| | - Ying Huang
- Clinical Molecular Medical Detection Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kuai Yu
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China; Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Pathology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Juan He
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China; Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Zhao
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China; Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Xie
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China; Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Ultrasound, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Li
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing, China; Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, China; Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
32
|
McManus M, Frangoul H, Steinberg MH. CRISPR-based gene therapy for the induction of fetal hemoglobin in sickle cell disease. Expert Rev Hematol 2024; 17:957-966. [PMID: 39535263 DOI: 10.1080/17474086.2024.2429605] [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: 06/12/2024] [Revised: 09/17/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Sickle cell disease is ameliorated and perhaps can be 'cured' if enough fetal hemoglobin is present in most erythrocytes. Hydroxyurea, which increases fetal hemoglobin levels, is widely available and effective, especially in children. Nevertheless, only cell-based gene therapy can achieve a 'curative' fetal hemoglobin threshold. AREAS COVERED We cover the path to modulating fetal hemoglobin gene expression and the use of CRISPR/Cas9 gene editing as a viable clinical modality for treating severe sickle cell disease relying on references obtained from PubMed. Mobilized autologous hematopoietic stem and progenitor cells are engineered with vectors that derepress genes that regulate fetal hemoglobin gene expression. Following myeloablative conditioning, gene-edited cells are reinfused, engrafted, and make large amounts of fetal hemoglobin. Within months, fetal hemoglobin forms more than 40% of the total hemoglobin and hemoglobin levels normalize; symptoms of sickle cell disease disappear. EXPERT OPINION Optimistically, these patients are 'cured,' but long term follow-up is needed. Although approved by regulatory agencies and highly efficacious, because of its technical imperatives and cost, this first gene editing therapeutic will be unavailable to most people with severe sickle cell disease. It is highly likely that improved methods of genomic editing will simplify gene therapy, reduce its costs, and lead to its wider applicability.
Collapse
Affiliation(s)
- Meghann McManus
- Sarah Cannon Pediatric Hematology/Oncology & Cellular Therapy @TriStar Centennial, Nashville, TN, USA
| | - Haydar Frangoul
- Sarah Cannon Pediatric Hematology/Oncology & Cellular Therapy @TriStar Centennial, Nashville, TN, USA
| | - Martin H Steinberg
- Department of Medicine, Division of Hematology and Medical Oncology, Center of Excellence for Sickle Cell Disease, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| |
Collapse
|
33
|
Zheng G, Yin M, Mehta S, Chu IT, Wang S, AlShaye A, Drainville K, Buyanbat A, Bienfait F, Tenglin K, Zhu Q, Orkin SH. A tetramer of BCL11A is required for stable protein production and fetal hemoglobin silencing. Science 2024; 386:1010-1018. [PMID: 39607926 DOI: 10.1126/science.adp3025] [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: 03/22/2024] [Revised: 08/08/2024] [Accepted: 10/21/2024] [Indexed: 11/30/2024]
Abstract
Down-regulation of BCL11A protein reverses the fetal (HbF, α2γ2) to adult (HbA, α2β2) hemoglobin switch and is exploited in gene-based therapy for hemoglobin disorders. Because of reliance on ex vivo cell manipulation and marrow transplant, such therapies cannot lessen disease burden. To develop new small-molecule approaches, we investigated the state of BCL11A protein in erythroid cells. We report that tetramer formation mediated by a single zinc finger (ZnF0) is required for production of steady-state protein. Beyond its role in protein stability, the tetramer state is necessary for γ-globin gene repression, because an engineered monomer fails to engage a critical co-repressor complex. These aspects of BCL11A protein production identify tetramer formation as a vulnerability for HbF silencing and provide opportunities for drug discovery.
Collapse
Affiliation(s)
- Ge Zheng
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Maolu Yin
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stuti Mehta
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - I-Te Chu
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stacy Wang
- Lester Sue Smith Breast Center, Department of Human Molecular Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Alia AlShaye
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Kirstin Drainville
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Altantsetseg Buyanbat
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Frédérique Bienfait
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Karin Tenglin
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Qian Zhu
- Lester Sue Smith Breast Center, Department of Human Molecular Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Stuart H Orkin
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
34
|
Diamantidis MD, Ikonomou G, Argyrakouli I, Pantelidou D, Delicou S. Genetic Modifiers of Hemoglobin Expression from a Clinical Perspective in Hemoglobinopathy Patients with Beta Thalassemia and Sickle Cell Disease. Int J Mol Sci 2024; 25:11886. [PMID: 39595957 PMCID: PMC11593634 DOI: 10.3390/ijms252211886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Hemoglobinopathies, namely β-thalassemia and sickle cell disease (SCD), are hereditary diseases, characterized by molecular genetic aberrations in the beta chains of hemoglobin. These defects affect the normal production of hemoglobin with severe anemia due to less or no amount of beta globins in patients with β-thalassemia (quantitative disorder), while SCD is a serious disease in which a mutated form of hemoglobin distorts the red blood cells into a crescent shape at low oxygen levels (qualitative disorder). Despite the revolutionary progress in recent years with the approval of gene therapy and gene editing for specific patients, there is an unmet need for highlighting the mechanisms influencing hemoglobin production and for the development of novel drugs and targeted therapies. The identification of the transcription factors and other genetic modifiers of hemoglobin expression is of utmost importance for discovering novel therapeutic approaches for patients with hemoglobinopathies. The aim of this review is to describe these complex molecular mechanisms and pathways affecting hemoglobin expression and to highlight the relevant investigational approaches or pharmaceutical interventions focusing on restoring the hemoglobin normal function by linking the molecular background of the disease with the clinical perspective. All the associated drugs increasing the hemoglobin expression in patients with hemoglobinopathies, along with gene therapy and gene editing, are also discussed.
Collapse
Affiliation(s)
- Michael D. Diamantidis
- Department of Hematology, Thalassemia and Sickle Cell Disease Unit, General Hospital of Larissa, 41221 Larissa, Greece;
| | - Georgia Ikonomou
- Thalassemia and Sickle Cell Disease Prevention Unit, General Hospital of Larissa, 41221 Larissa, Greece;
| | - Ioanna Argyrakouli
- Department of Hematology, Thalassemia and Sickle Cell Disease Unit, General Hospital of Larissa, 41221 Larissa, Greece;
| | - Despoina Pantelidou
- Thalassemia and Sickle Cell Disease Unit, AHEPA University General Hospital, 41221 Thessaloniki, Greece;
| | - Sophia Delicou
- Center of Expertise in Hemoglobinopathies and Their Complications, Thalassemia and Sickle Cell Disease Unit, Hippokration General Hospital, 41221 Athens, Greece;
| |
Collapse
|
35
|
Olalla B, Río P. A new breakthrough in genome editing: the story of Casgevy. Cytotherapy 2024; 26:1299-1300. [PMID: 38944796 DOI: 10.1016/j.jcyt.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 07/01/2024]
Affiliation(s)
- Beatriz Olalla
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain; Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
| | - Paula Río
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid, Spain; Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain.
| |
Collapse
|
36
|
Muhtaseb AW, Duan J. Modeling common and rare genetic risk factors of neuropsychiatric disorders in human induced pluripotent stem cells. Schizophr Res 2024; 273:39-61. [PMID: 35459617 PMCID: PMC9735430 DOI: 10.1016/j.schres.2022.04.003] [Citation(s) in RCA: 7] [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: 02/14/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022]
Abstract
Recent genome-wide association studies (GWAS) and whole-exome sequencing of neuropsychiatric disorders, especially schizophrenia, have identified a plethora of common and rare disease risk variants/genes. Translating the mounting human genetic discoveries into novel disease biology and more tailored clinical treatments is tied to our ability to causally connect genetic risk variants to molecular and cellular phenotypes. When combined with the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated (Cas) nuclease-mediated genome editing system, human induced pluripotent stem cell (hiPSC)-derived neural cultures (both 2D and 3D organoids) provide a promising tractable cellular model for bridging the gap between genetic findings and disease biology. In this review, we first conceptualize the advances in understanding the disease polygenicity and convergence from the past decade of iPSC modeling of different types of genetic risk factors of neuropsychiatric disorders. We then discuss the major cell types and cellular phenotypes that are most relevant to neuropsychiatric disorders in iPSC modeling. Finally, we critically review the limitations of iPSC modeling of neuropsychiatric disorders and outline the need for implementing and developing novel methods to scale up the number of iPSC lines and disease risk variants in a systematic manner. Sufficiently scaled-up iPSC modeling and a better functional interpretation of genetic risk variants, in combination with cutting-edge CRISPR/Cas9 gene editing and single-cell multi-omics methods, will enable the field to identify the specific and convergent molecular and cellular phenotypes in precision for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Abdurrahman W Muhtaseb
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, United States of America; Department of Human Genetics, The University of Chicago, Chicago, IL 60637, United States of America
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, United States of America; Department of Psychiatry and Behavioral Neuroscience, The University of Chicago, Chicago, IL 60637, United States of America.
| |
Collapse
|
37
|
Wang Y, Hon GC. Towards functional maps of non-coding variants in cancer. Front Genome Ed 2024; 6:1481443. [PMID: 39544254 PMCID: PMC11560456 DOI: 10.3389/fgeed.2024.1481443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
Large scale cancer genomic studies in patients have unveiled millions of non-coding variants. While a handful have been shown to drive cancer development, the vast majority have unknown function. This review describes the challenges of functionally annotating non-coding cancer variants and understanding how they contribute to cancer. We summarize recently developed high-throughput technologies to address these challenges. Finally, we outline future prospects for non-coding cancer genetics to help catalyze personalized cancer therapy.
Collapse
Affiliation(s)
- Yihan Wang
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Gary C. Hon
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Division of Basic Reproductive Biology Research, Department of Obstetrics and Gynecology, Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
38
|
Lessard S, Rimmelé P, Ling H, Moran K, Vieira B, Lin YD, Rajani GM, Hong V, Reik A, Boismenu R, Hsu B, Chen M, Cockroft BM, Uchida N, Tisdale J, Alavi A, Krishnamurti L, Abedi M, Galeon I, Reiner D, Wang L, Ramezi A, Rendo P, Walters MC, Levasseur D, Peters R, Harris T, Hicks A. Zinc finger nuclease-mediated gene editing in hematopoietic stem cells results in reactivation of fetal hemoglobin in sickle cell disease. Sci Rep 2024; 14:24298. [PMID: 39414860 PMCID: PMC11484757 DOI: 10.1038/s41598-024-74716-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 09/27/2024] [Indexed: 10/18/2024] Open
Abstract
BIVV003 is a gene-edited autologous cell therapy in clinical development for the potential treatment of sickle cell disease (SCD). Hematopoietic stem cells (HSC) are genetically modified with mRNA encoding zinc finger nucleases (ZFN) that target and disrupt a specific regulatory GATAA motif in the BCL11A erythroid enhancer to reactivate fetal hemoglobin (HbF). We characterized ZFN-edited HSC from healthy donors and donors with SCD. Results of preclinical studies show that ZFN-mediated editing is highly efficient, with enriched biallelic editing and high frequency of on-target indels, producing HSC capable of long-term multilineage engraftment in vivo, and express HbF in erythroid progeny. Interim results from the Phase 1/2 PRECIZN-1 study demonstrated that BIVV003 was well-tolerated in seven participants with SCD, of whom five of the six with more than 3 months of follow-up displayed increased total hemoglobin and HbF, and no severe vaso-occlusive crises. Our data suggest BIVV003 represents a compelling and novel cell therapy for the potential treatment of SCD.
Collapse
Affiliation(s)
- Samuel Lessard
- Rare Blood Disorders, Sanofi, Waltham, MA, 02451, USA.
- Precision Medicine and Computational Biology, Sanofi, Cambridge, MA, 02141, USA.
| | | | - Hui Ling
- Rare Blood Disorders, Sanofi, Waltham, MA, 02451, USA
| | - Kevin Moran
- Rare Blood Disorders, Sanofi, Waltham, MA, 02451, USA
| | | | - Yi-Dong Lin
- Rare Blood Disorders, Sanofi, Waltham, MA, 02451, USA
| | | | - Vu Hong
- Rare Blood Disorders, Sanofi, Waltham, MA, 02451, USA
| | | | | | - Ben Hsu
- Sangamo Therapeutics, Richmond, CA, 94804, USA
| | | | | | - Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes/National Institute of Diabetes and Digestive and Kidney Diseases, National Heart, National Institutes of Health (NIH), Bethesda, MD, USA
| | - John Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institutes/National Institute of Diabetes and Digestive and Kidney Diseases, National Heart, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Asif Alavi
- Henry Ford Cancer Institute, Detroit, MI, USA
| | - Lakshmanan Krishnamurti
- Emory University, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Mehrdad Abedi
- University of California-Davis Medical Center, Sacramento, CA, USA
| | | | - David Reiner
- Rare Blood Disorders, Sanofi, Waltham, MA, 02451, USA
| | - Lin Wang
- Rare Blood Disorders, Sanofi, Waltham, MA, 02451, USA
| | - Anne Ramezi
- Rare Blood Disorders, Sanofi, Waltham, MA, 02451, USA
| | - Pablo Rendo
- Rare Blood Disorders, Sanofi, Waltham, MA, 02451, USA
| | - Mark C Walters
- University of California San Francisco Benioff Children's Hospital, Oakland, CA, USA
| | | | - Robert Peters
- Rare Blood Disorders, Sanofi, Waltham, MA, 02451, USA
| | | | - Alexandra Hicks
- Rare Blood Disorders, Sanofi, Waltham, MA, 02451, USA
- Immunology and Inflammation, Sanofi, Cambridge, MA, 02141, USA
| |
Collapse
|
39
|
Rivest JF, Carter S, Goupil C, Antérieux P, Cyr D, Ung RV, Dal Soglio D, Mac-Way F, Waters PJ, Paganelli M, Doyon Y. In vivo dissection of the mouse tyrosine catabolic pathway with CRISPR-Cas9 identifies modifier genes affecting hereditary tyrosinemia type 1. Genetics 2024; 228:iyae139. [PMID: 39178380 PMCID: PMC11457941 DOI: 10.1093/genetics/iyae139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 08/12/2024] [Indexed: 08/25/2024] Open
Abstract
Hereditary tyrosinemia type 1 is an autosomal recessive disorder caused by mutations (pathogenic variants) in fumarylacetoacetate hydrolase, an enzyme involved in tyrosine degradation. Its loss results in the accumulation of toxic metabolites that mainly affect the liver and kidneys and can lead to severe liver disease and liver cancer. Tyrosinemia type 1 has a global prevalence of approximately 1 in 100,000 births but can reach up to 1 in 1,500 births in some regions of Québec, Canada. Mutating functionally related "modifier' genes (i.e. genes that, when mutated, affect the phenotypic impacts of mutations in other genes) is an emerging strategy for treating human genetic diseases. In vivo somatic genome editing in animal models of these diseases is a powerful means to identify modifier genes and fuel treatment development. In this study, we demonstrate that mutating additional enzymes in the tyrosine catabolic pathway through liver-specific genome editing can relieve or worsen the phenotypic severity of a murine model of tyrosinemia type 1. Neonatal gene delivery using recombinant adeno-associated viral vectors expressing Staphylococcus aureus Cas9 under the control of a liver-specific promoter led to efficient gene disruption and metabolic rewiring of the pathway, with systemic effects that were distinct from the phenotypes observed in whole-body knockout models. Our work illustrates the value of using in vivo genome editing in model organisms to study the direct effects of combining pathological mutations with modifier gene mutations in isogenic settings.
Collapse
Affiliation(s)
- Jean-François Rivest
- Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine, Laval University, Québec City, QC G1V 4G2, Canada
- Université Laval Cancer Research Centre, Québec City, QC G1V 0A6, Canada
| | - Sophie Carter
- Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine, Laval University, Québec City, QC G1V 4G2, Canada
- Université Laval Cancer Research Centre, Québec City, QC G1V 0A6, Canada
| | - Claudia Goupil
- Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine, Laval University, Québec City, QC G1V 4G2, Canada
- Université Laval Cancer Research Centre, Québec City, QC G1V 0A6, Canada
| | - Pénélope Antérieux
- Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine, Laval University, Québec City, QC G1V 4G2, Canada
- Université Laval Cancer Research Centre, Québec City, QC G1V 0A6, Canada
| | - Denis Cyr
- Medical Genetics Service, Dept. Laboratory Medicine and Dept. Pediatrics, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC J1H 5N4, Canada
| | - Roth-Visal Ung
- Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine, Laval University, Québec City, QC G1V 4G2, Canada
| | - Dorothée Dal Soglio
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - Fabrice Mac-Way
- Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine, Laval University, Québec City, QC G1V 4G2, Canada
| | - Paula J Waters
- Medical Genetics Service, Dept. Laboratory Medicine and Dept. Pediatrics, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC J1H 5N4, Canada
| | - Massimiliano Paganelli
- Centre Hospitalier Universitaire Sainte-Justine Research Center, Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - Yannick Doyon
- Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine, Laval University, Québec City, QC G1V 4G2, Canada
- Université Laval Cancer Research Centre, Québec City, QC G1V 0A6, Canada
| |
Collapse
|
40
|
Martyn GE, Doerfler PA, Yao Y, Quinlan KGR, Weiss MJ, Crossley M. Hydroxyurea reduces the levels of the fetal globin gene repressors ZBTB7A/LRF and BCL11A in erythroid cells in vitro. JOURNAL OF SICKLE CELL DISEASE 2024; 1:yoae008. [PMID: 40304012 PMCID: PMC12039817 DOI: 10.1093/jscdis/yoae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/06/2024] [Accepted: 09/03/2024] [Indexed: 05/02/2025]
Abstract
Objectives Hydroxyurea (HU) is the most widely used therapy for adults and children with sickle cell disease (SCD). It is believed to act largely by inducing the transcription of fetal γ-globin genes to generate fetal hemoglobin (HbF), which inhibits the pathological polymerization of sickle hemoglobin (HbS). The mechanisms by which hydroxyurea elevates HbF are unclear. We explored the hypothesis that hydroxyurea induces HbF expression by inhibiting the expression of 2 γ-globin gene repressors, BCL11A and ZBTB7A (also known as LRF), which normally bind the γ-globin gene promoters to inhibit their expression after birth. Methods We treated immortalized murine erythroleukemia cells and normal human donor CD34+ hematopoietic stem and progenitor cell-derived erythroblasts with hydroxyurea and measured the effects on globin, BCL11A and ZBTB7A protein and mRNA expression. Results Treating murine erythroleukemia cells or human CD34+ hematopoietic stem and progenitor cell-derived erythroblasts with hydroxyurea reduced the protein levels of BCL11A and ZBTB7A compared to the vehicle-treated control. BCL11A mRNA levels were reduced in both cell types upon hydroxyurea treatment. However, ZBTB7A mRNA levels were only reduced in human CD34+ hematopoietic stem and progenitor cell-derived erythroblasts. Conclusions Hydroxyurea can act in erythroid cells to reduce the levels and activity of two direct fetal γ-globin transcriptional repressors with accompanying de-repression of the γ-globin genes and induction of HbF, which may explain the mechanism of action leading to amelioration of symptoms in SCD patients treated with this drug.
Collapse
Affiliation(s)
- Gabriella E Martyn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW) Sydney, Sydney, NSW 2052, Australia
| | - Phillip A Doerfler
- Department of Hematology, St Jude Children’s Research Hospital, Memphis, TN 38105-3678, United States
| | - Yu Yao
- Department of Hematology, St Jude Children’s Research Hospital, Memphis, TN 38105-3678, United States
| | - Kate G R Quinlan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW) Sydney, Sydney, NSW 2052, Australia
| | - Mitchell J Weiss
- Department of Hematology, St Jude Children’s Research Hospital, Memphis, TN 38105-3678, United States
| | - Merlin Crossley
- School of Biotechnology and Biomolecular Sciences, University of New South Wales (UNSW) Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
41
|
Weaver SB, Singh D, Wilson KM. Gene Therapies for Sickle Cell Disease. J Pharm Technol 2024; 40:236-247. [PMID: 39391326 PMCID: PMC11463071 DOI: 10.1177/87551225241268742] [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: 10/12/2024] Open
Abstract
Background: Sickle cell disease (SCD) is a prevalent autosomal recessive hemoglobinopathy affecting millions worldwide, particularly individuals of African ancestry. Sickle cell disease is a lifelong condition associated with a negative impact on quality of life and mortality, causing complications such as painful vaso-occlusive episodes, acute chest syndrome, stroke, long-term anemia, and end-organ damage. Currently, there are 4 U.S. Food and Drug Administration (FDA)-approved drugs, including hydroxyurea, l-glutamine, voxelotor, and crizanlizumab, which work to alleviate symptoms and prevent complications associated with SCD, albeit without addressing the underlying cause of SCD. Allogeneic hematopoietic stem cell transplant (HSCT) has shown promise as a curative approach to SCD but is limited by donor availability and associated complications. This paper aims to review the efficacy and safety of exagamglogene autotemcel and lovotibeglogene autotemcel for managing patients with SCD, including their place in therapy, cost, and accessibility in clinical care. Data Sources: The authors searched PubMed and Medline from 2017 to 2024, for primary literature on both exagamglogene autotemcel and lovotibeglogene autotemcel. Results: The authors identified relevant studies and summarized the data on the two gene therapies. Conclusion: Exagamglogene autotemcel and lovotibeglogene autotemcel are two management strategies that address the underlying cause of SCD and provide curative potential for patients with SCD.
Collapse
Affiliation(s)
| | - Divita Singh
- Temple University School of Pharmacy, Philadelphia, PA, USA
| | | |
Collapse
|
42
|
Bresnick EH, Papayannopoulou T, Migliaccio AR. Mechanistic and Biological Perspectives on Erythropoiesis. Exp Hematol 2024; 138:104286. [PMID: 39034024 DOI: 10.1016/j.exphem.2024.104286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Affiliation(s)
- Emery H Bresnick
- Wisconsin Blood Cancer Research Institute, Carbone Cancer Center, Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | | | - Anna Rita Migliaccio
- Altius Institute for Biomedical Sciences, Seattle, WA, USA; Institute of Nanotechnology, National Research Council (Cnr-NANOTEC), c/o Campus Ecotekne, Lecce, Italy
| |
Collapse
|
43
|
George CA, Sahu SU, de Oñate L, Souza BSDF, Wilson RC. Genome Editing Therapy for the Blood: Ex Vivo Success and In Vivo Prospects. CRISPR J 2024; 7:231-248. [PMID: 39324895 DOI: 10.1089/crispr.2024.0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024] Open
Abstract
Hematopoietic stem cells (HSCs) provide the body with a continuous supply of healthy, functional blood cells. In patients with hematopoietic malignancies, immunodeficiencies, lysosomal storage disorders, and hemoglobinopathies, therapeutic genome editing offers hope for corrective intervention, with even modest editing efficiencies likely to provide clinical benefit. Engineered white blood cells, such as T cells, can be applied therapeutically to address monogenic disorders of the immune system, HIV infection, or cancer. The versatility of CRISPR-based tools allows countless new medical interventions for diseases of the blood, and rapid ex vivo success has been demonstrated in hemoglobinopathies via transplantation of the patient's HSCs following genome editing in a laboratory setting. Here we review recent advances in therapeutic genome editing of HSCs and T cells, focusing on the progress in ex vivo contexts, the promise of improved access via in vivo delivery, as well as the ongoing preclinical efforts that may enable the transition from ex vivo to in vivo administration. We discuss the challenges, limitations, and future prospects of this rapidly developing field, which may one day establish CRISPR as the standard of care for some diseases affecting the blood.
Collapse
Affiliation(s)
- Christy A George
- Innovative Genomics Institute, University of California Berkeley, Berkeley, California, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, USA
| | - Srishti U Sahu
- Innovative Genomics Institute, University of California Berkeley, Berkeley, California, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, USA
| | - Lorena de Oñate
- Innovative Genomics Institute, University of California Berkeley, Berkeley, California, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, USA
| | - Bruno Solano de Freitas Souza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- Pioneer Science Initiative, D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Ross C Wilson
- Innovative Genomics Institute, University of California Berkeley, Berkeley, California, USA
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California, USA
- California Institute for Quantitative Biosciences, University of California Berkeley, Berkeley, California, USA
| |
Collapse
|
44
|
Martin-Rufino JD, Caulier A, Lee S, Castano N, King E, Joubran S, Jones M, Goldman SR, Arora UP, Wahlster L, Lander ES, Sankaran VG. Transcription factor networks disproportionately enrich for heritability of blood cell phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.611392. [PMID: 39314298 PMCID: PMC11419094 DOI: 10.1101/2024.09.09.611392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Most phenotype-associated genetic variants map to non-coding regulatory regions of the human genome. Moreover, variants associated with blood cell phenotypes are enriched in regulatory regions active during hematopoiesis. To systematically explore the nature of these regions, we developed a highly efficient strategy, Perturb-multiome, that makes it possible to simultaneously profile both chromatin accessibility and gene expression in single cells with CRISPR-mediated perturbation of a range of master transcription factors (TFs). This approach allowed us to examine the connection between TFs, accessible regions, and gene expression across the genome throughout hematopoietic differentiation. We discovered that variants within the TF-sensitive accessible chromatin regions, while representing less than 0.3% of the genome, show a ~100-fold enrichment in heritability across certain blood cell phenotypes; this enrichment is strikingly higher than for other accessible chromatin regions. Our approach facilitates large-scale mechanistic understanding of phenotype-associated genetic variants by connecting key cis-regulatory elements and their target genes within gene regulatory networks.
Collapse
Affiliation(s)
- Jorge Diego Martin-Rufino
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Equally contributed to work
| | - Alexis Caulier
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Equally contributed to work
| | - Seayoung Lee
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Equally contributed to work
| | - Nicole Castano
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Equally contributed to work
| | - Emily King
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Samantha Joubran
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Marcus Jones
- Nascent Transcriptomics Core, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Seth R. Goldman
- Nascent Transcriptomics Core, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Uma P. Arora
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Lara Wahlster
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Eric S. Lander
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Vijay G. Sankaran
- Division of Hematology/Oncology, Boston Children’s Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| |
Collapse
|
45
|
Tanshee RR, Mahmud Z, Nabi AHMN, Sayem M. A comprehensive in silico investigation into the pathogenic SNPs in the RTEL1 gene and their biological consequences. PLoS One 2024; 19:e0309713. [PMID: 39240887 PMCID: PMC11379182 DOI: 10.1371/journal.pone.0309713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 08/16/2024] [Indexed: 09/08/2024] Open
Abstract
The Regulator of Telomere Helicase 1 (RTEL1) gene encodes a critical DNA helicase intricately involved in the maintenance of telomeric structures and the preservation of genomic stability. Germline mutations in the RTEL1 gene have been clinically associated with Hoyeraal-Hreidarsson syndrome, a more severe version of Dyskeratosis Congenita. Although various research has sought to link RTEL1 mutations to specific disorders, no comprehensive investigation has yet been conducted on missense mutations. In this study, we attempted to investigate the functionally and structurally deleterious coding and non-coding SNPs of the RTEL1 gene using an in silico approach. Initially, out of 1392 nsSNPs, 43 nsSNPs were filtered out through ten web-based bioinformatics tools. With subsequent analysis using nine in silico tools, these 43 nsSNPs were further shortened to 11 most deleterious nsSNPs. Furthermore, analyses of mutated protein structures, evolutionary conservancy, surface accessibility, domains & PTM sites, cancer susceptibility, and interatomic interaction revealed the detrimental effect of these 11 nsSNPs on RTEL1 protein. An in-depth investigation through molecular docking with the DNA binding sequence demonstrated a striking change in the interaction pattern for F15L, M25V, and G706R mutant proteins, suggesting the more severe consequences of these mutations on protein structure and functionality. Among the non-coding variants, two had the highest likelihood of being regulatory variants, whereas one variant was predicted to affect the target region of a miRNA. Thus, this study lays the groundwork for extensive analysis of RTEL1 gene variants in the future, along with the advancement of precision medicine and other treatment modalities.
Collapse
Affiliation(s)
- Rifah Rownak Tanshee
- Department of Mathematics and Natural Sciences, BRAC University, Badda, Dhaka, Bangladesh
| | - Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - A. H. M. Nurun Nabi
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - Mohammad Sayem
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
46
|
Testa U, Leone G, Cappellini MD. Therapeutic Gene Editing for Hemoglobinopathies. Mediterr J Hematol Infect Dis 2024; 16:e2024068. [PMID: 39258178 PMCID: PMC11385271 DOI: 10.4084/mjhid.2024.068] [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: 07/13/2024] [Accepted: 08/07/2024] [Indexed: 09/12/2024] Open
Abstract
In the last ten years, a consistent number of clinical studies have evaluated different gene approaches for the treatment of patients with sickle cell disease (SCD) and transfusion-dependent β-thalassemia (TDT). Initial studies of gene therapy for hemoglobinopathies involved the use of lentiviral vectors to add functional copies of the gene encoding β-globin in defective CD34 cells; more recently, gene editing techniques have been used involving either CRISPR-Cas9, transcription activation-like effector protein nuclease, zinc finger nuclease, and base editing to either induce fetal hemoglobin production at therapeutic levels or to genetically repair the underlying molecular defect causing the disease. Here, we review recent gene editing studies that have started the development of a new era in the treatment of hemoglobinopathies and, in general, monoallelic hereditary diseases.
Collapse
Affiliation(s)
- Ugo Testa
- Istituto Superiore Sanità, Roma, Italy
| | - Giuseppe Leone
- Department of Radiological and Hematological Sciences, Catholic University, Rome, Italy
| | - Maria Domenica Cappellini
- Department of Clinical Sciences and Community, University of Milan, IRCCS Ca' Granda Foundation Maggiore Policlinico Hospital, Milan, Italy
| |
Collapse
|
47
|
Xiang G, He X, Giardine BM, Isaac KJ, Taylor DJ, McCoy RC, Jansen C, Keller CA, Wixom AQ, Cockburn A, Miller A, Qi Q, He Y, Li Y, Lichtenberg J, Heuston EF, Anderson SM, Luan J, Vermunt MW, Yue F, Sauria MEG, Schatz MC, Taylor J, Göttgens B, Hughes JR, Higgs DR, Weiss MJ, Cheng Y, Blobel GA, Bodine DM, Zhang Y, Li Q, Mahony S, Hardison RC. Interspecies regulatory landscapes and elements revealed by novel joint systematic integration of human and mouse blood cell epigenomes. Genome Res 2024; 34:1089-1105. [PMID: 38951027 PMCID: PMC11368181 DOI: 10.1101/gr.277950.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
Knowledge of locations and activities of cis-regulatory elements (CREs) is needed to decipher basic mechanisms of gene regulation and to understand the impact of genetic variants on complex traits. Previous studies identified candidate CREs (cCREs) using epigenetic features in one species, making comparisons difficult between species. In contrast, we conducted an interspecies study defining epigenetic states and identifying cCREs in blood cell types to generate regulatory maps that are comparable between species, using integrative modeling of eight epigenetic features jointly in human and mouse in our Validated Systematic Integration (VISION) Project. The resulting catalogs of cCREs are useful resources for further studies of gene regulation in blood cells, indicated by high overlap with known functional elements and strong enrichment for human genetic variants associated with blood cell phenotypes. The contribution of each epigenetic state in cCREs to gene regulation, inferred from a multivariate regression, was used to estimate epigenetic state regulatory potential (esRP) scores for each cCRE in each cell type, which were used to categorize dynamic changes in cCREs. Groups of cCREs displaying similar patterns of regulatory activity in human and mouse cell types, obtained by joint clustering on esRP scores, harbor distinctive transcription factor binding motifs that are similar between species. An interspecies comparison of cCREs revealed both conserved and species-specific patterns of epigenetic evolution. Finally, we show that comparisons of the epigenetic landscape between species can reveal elements with similar roles in regulation, even in the absence of genomic sequence alignment.
Collapse
Affiliation(s)
- Guanjue Xiang
- Bioinformatics and Genomics Graduate Program, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02215, USA
| | - Xi He
- Bioinformatics and Genomics Graduate Program, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Belinda M Giardine
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Kathryn J Isaac
- Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Dylan J Taylor
- Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Rajiv C McCoy
- Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Camden Jansen
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Cheryl A Keller
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Alexander Q Wixom
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - April Cockburn
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Amber Miller
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Qian Qi
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Yanghua He
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaìi at Mānoa, Honolulu, Hawaii 96822, USA
| | - Yichao Li
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Jens Lichtenberg
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, Bethesda, Maryland 20892, USA
| | - Elisabeth F Heuston
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, Bethesda, Maryland 20892, USA
| | - Stacie M Anderson
- Flow Cytometry Core, National Human Genome Research Institute, Bethesda, Maryland 20892, USA
| | - Jing Luan
- Department of Pediatrics, Children's Hospital of Philadelphia, and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Marit W Vermunt
- Department of Pediatrics, Children's Hospital of Philadelphia, and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Feng Yue
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60611, USA
| | - Michael E G Sauria
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Michael C Schatz
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - James Taylor
- Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Berthold Göttgens
- Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, United Kingdom
| | - Jim R Hughes
- MRC Weatherall Institute of Molecular Medicine, Oxford University, Oxford OX3 9DS, United Kingdom
| | - Douglas R Higgs
- MRC Weatherall Institute of Molecular Medicine, Oxford University, Oxford OX3 9DS, United Kingdom
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Yong Cheng
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Gerd A Blobel
- Department of Pediatrics, Children's Hospital of Philadelphia, and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - David M Bodine
- Genetics and Molecular Biology Branch, National Human Genome Research Institute, Bethesda, Maryland 20892, USA
| | - Yu Zhang
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Qunhua Li
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
- Center for Computational Biology and Bioinformatics, Genome Sciences Institute, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Shaun Mahony
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
- Center for Computational Biology and Bioinformatics, Genome Sciences Institute, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Ross C Hardison
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania 16802, USA;
- Center for Computational Biology and Bioinformatics, Genome Sciences Institute, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| |
Collapse
|
48
|
Lin J, Nguyen MA, Lin LY, Zeng J, Verma A, Neri NR, da Silva LF, Mucci A, Wolfe S, Shaw KL, Clement K, Brendel C, Pinello L, Pellin D, Bauer DE. Scalable assessment of genome editing off-targets associated with genetic variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.605019. [PMID: 39211178 PMCID: PMC11360989 DOI: 10.1101/2024.07.24.605019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Genome editing with RNA-guided DNA binding factors carries risk of off-target editing at homologous sequences. Genetic variants may introduce sequence changes that increase homology to a genome editing target, thereby increasing risk of off-target editing. Conventional methods to verify candidate off-targets rely on access to cells with genomic DNA carrying these sequences. However, for candidate off-targets associated with genetic variants, appropriate cells for experimental verification may not be available. Here we develop a method, Assessment By Stand-in Off-target LentiViral Ensemble with sequencing (ABSOLVE-seq), to integrate a set of candidate off-target sequences along with unique molecular identifiers (UMIs) in genomes of primary cells followed by clinically relevant gene editor delivery. Gene editing of dozens of candidate off-target sequences may be evaluated in a single experiment with high sensitivity, precision, and power. We provide an open-source pipeline to analyze sequencing data. This approach enables experimental assessment of the influence of human genetic diversity on specificity evaluation during gene editing therapy development.
Collapse
|
49
|
Higgs D, Kassouf M. Developing a pill to treat sickle cell disease. Science 2024; 385:27-28. [PMID: 38963861 DOI: 10.1126/science.adq3757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
A newly identified epigenetic modifier increases fetal hemoglobin in preclinical studies.
Collapse
Affiliation(s)
- Douglas Higgs
- Laboratory of Gene Regulation, MRC Weatherall Institute of Molecular Medicine, and Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| | - Mira Kassouf
- Laboratory of Gene Regulation, MRC Weatherall Institute of Molecular Medicine, and Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK
| |
Collapse
|
50
|
Iñiguez-Muñoz S, Llinàs-Arias P, Ensenyat-Mendez M, Bedoya-López AF, Orozco JIJ, Cortés J, Roy A, Forsberg-Nilsson K, DiNome ML, Marzese DM. Hidden secrets of the cancer genome: unlocking the impact of non-coding mutations in gene regulatory elements. Cell Mol Life Sci 2024; 81:274. [PMID: 38902506 PMCID: PMC11335195 DOI: 10.1007/s00018-024-05314-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 12/07/2023] [Accepted: 06/06/2024] [Indexed: 06/22/2024]
Abstract
Discoveries in the field of genomics have revealed that non-coding genomic regions are not merely "junk DNA", but rather comprise critical elements involved in gene expression. These gene regulatory elements (GREs) include enhancers, insulators, silencers, and gene promoters. Notably, new evidence shows how mutations within these regions substantially influence gene expression programs, especially in the context of cancer. Advances in high-throughput sequencing technologies have accelerated the identification of somatic and germline single nucleotide mutations in non-coding genomic regions. This review provides an overview of somatic and germline non-coding single nucleotide alterations affecting transcription factor binding sites in GREs, specifically involved in cancer biology. It also summarizes the technologies available for exploring GREs and the challenges associated with studying and characterizing non-coding single nucleotide mutations. Understanding the role of GRE alterations in cancer is essential for improving diagnostic and prognostic capabilities in the precision medicine era, leading to enhanced patient-centered clinical outcomes.
Collapse
Affiliation(s)
- Sandra Iñiguez-Muñoz
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Pere Llinàs-Arias
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Miquel Ensenyat-Mendez
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Andrés F Bedoya-López
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Javier I J Orozco
- Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, 08017, Barcelona, Spain
- Medica Scientia Innovation Research SL (MEDSIR), 08018, Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, 28670, Madrid, Spain
| | - Ananya Roy
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Maggie L DiNome
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Diego M Marzese
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain.
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|