1
|
Labouta HI. Defining the landscape of prenatal nanomedicine and a roadmap for future research. Nanomedicine (Lond) 2025:1-5. [PMID: 40249012 DOI: 10.1080/17435889.2025.2492540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Affiliation(s)
- Hagar I Labouta
- The Keenan Research Centre for Biomedical Science, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
Kolarova TR, Cheng E. In Utero Therapies, the Next Frontier. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2025:e32135. [PMID: 40126136 DOI: 10.1002/ajmg.c.32135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 03/25/2025]
Abstract
Advancements in molecular diagnostics and the expanded understanding of pathophysiologic processes underlying a variety of genetic conditions have led to the design and implementation of many targeted therapies in the past decade. In the prenatal space, these advancements have rapidly changed the field of prenatal diagnosis and have garnered enthusiasm toward interventions aimed at correcting specific disease mechanisms in utero to prevent irreversible injury and/or leverage fetal physiology to increase the effectiveness of these treatments. Although many promising trials are underway, continued efforts to fully elucidate the intricacies of fetal immunology, the fetal blood-brain barrier and precise molecular processes during different stages of development will be paramount to continued refinement of in utero therapies. The social and ethical implications of fetal therapy are also of utmost importance, especially in an era of increasing restrictions on reproductive autonomy. This review aims to summarize current efforts toward in utero targeted fetal therapies as well as the history and ethical implications of such endeavors.
Collapse
Affiliation(s)
- Teodora R Kolarova
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Edith Cheng
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
- Division of Medical Genetics, Department of Internal Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
3
|
Spagnolo P, Tonelli R, Mura M, Reisman W, Sotiropoulou V, Tzouvelekis A. Investigational gene expression inhibitors for the treatment of idiopathic pulmonary fibrosis. Expert Opin Investig Drugs 2025; 34:61-80. [PMID: 39916340 DOI: 10.1080/13543784.2025.2462592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 01/31/2025] [Indexed: 02/12/2025]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive fibrosing interstitial lung disease of unknown cause that occurs primarily in older adults and is associated with poor quality of life and substantial healthcare utilization. IPF has a dismal prognosis. Indeed, first-line therapy, which includes nintedanib and pirfenidone, does not stop disease progression and is often associated with tolerability issues. Therefore, there remains a high medical need for more efficacious and better tolerated treatments. AREAS COVERED Gene therapy is a relatively unexplored field of research in IPF that has the potential to mitigate a range of profibrotic pathways by introducing genetic material into cells. Here, we summarize and critically discuss publications that have explored the safety and efficacy of gene therapy in experimentally-induced pulmonary fibrosis in animals, as clinical studies in humans have not been published yet. EXPERT OPINION The application of gene therapy in pulmonary fibrosis requires further investigation to address several technical and biological hurdles, improve vectors' design, drug delivery, and target selection, mitigate off-target effects and develop markers of gene penetration into target cells. Long-term clinical data are needed to bring gene therapy in IPF one step closer to practice.
Collapse
Affiliation(s)
- Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Roberto Tonelli
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University Hospital of Modena and Reggio Emilia, Modena, Italy
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Modena, Italy
| | - Marco Mura
- Division of Respirology, Western University, London, Ontario, Canada
| | - William Reisman
- Division of Respirology, Western University, London, Ontario, Canada
| | | | - Argyrios Tzouvelekis
- Department of Respiratory Medicine, University Hospital of Patras, Patras, Greece
| |
Collapse
|
4
|
Habib I, Jawed JJ, Nasrin T, Shaikh S. Briefing of pulmonary sarcoidosis: Reduction-oxidation, misleading and possibilities. Indian J Tuberc 2025; 72:103-111. [PMID: 39890360 DOI: 10.1016/j.ijtb.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 02/03/2025]
Abstract
Sarcoidosis is an inflammatory disease with limited treatment strategies and is characterized by the presence of abnormal lumps (granulomas) of the inflammatory cells. Among the types, pulmonary sarcoidosis most commonly occurs (about 90%), affecting the lungs and intrathoracic lymph nodes. Although the cause of its occurrence is still unknown, perhaps microbes and chemical exposures, as well as genetic history, may trigger the disease occurrence. The updated scenario also depicted the interconnection between oxidative stress and pulmonary sarcoidosis. Thus, the therapeutic value of the genetic consequences, as well as the redox status of pulmonary sarcoidosis, are under consideration. In addition, sarcoidosis complexity has been associated with tumor malignancy and tuberculosis. Therefore, in this review, we summarized the current status of pulmonary sarcoidosis, interference of lung cancer and tuberculosis complications, understanding of the role of reactive species in disease occurrence, and how they are associated with genetic features.
Collapse
Affiliation(s)
- Irfan Habib
- Department of Internal Medicine, College of Medicine and JNM Hospital, WB, India
| | - Junaid Jibran Jawed
- Institute of Health Sciences, Presidency University-2nd Campus, DG/02/02, New Town, Rajarhat, Kolkata, 700156, India
| | - Tina Nasrin
- Dept. of Electrical and Computer Engineering, University of Cyprus, Nicosia, Cyprus
| | - Soni Shaikh
- Laboratory of Histopathology, TATA MEDICAL CENTER 14, MAR (E-W), New Town, Rajarhat, Kolkata, 700160, India.
| |
Collapse
|
5
|
Eaton DA, Lynn AY, Surprenant JM, Deschenes EI, Guerra ME, Rivero R, Yung NK, O’Connor M, Glazer PM, Bahtiyar MO, Saltzman WM, Stitelman DH. Biodistribution of Polymeric Nanoparticles following in utero Delivery to a Nonhuman Primate. Biomed Hub 2025; 10:23-32. [PMID: 39845408 PMCID: PMC11753793 DOI: 10.1159/000543138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Monogenic diseases can be diagnosed before birth. Systemic fetal administration of nanoparticles (NPs) grants therapeutic access to developing stem cell populations impacted by these classes of disease. Delivery of editing reagents in these NPs administered before birth has yielded encouraging results in preclinical mouse models of monogenic diseases. Methods To translate this strategy clinically, the safety and efficacy of this strategy in larger animals will be necessary. We performed a pilot biodistribution study in 3 fetal nonhuman primates (NHPs) in mid-gestation examining systemic delivery of polymeric NPs loaded with fluorescent dye. Results We found several similarities in distribution to our experience in mice, namely, extensive uptake in fetal liver and spleen. A striking finding that is not recapitulated in the mouse was the accumulation of NPs in the zones of proliferation and ossification of the fetal bone. Of great importance, there did not appear to be NP accumulation in the fetal male or female germline zones or maternal tissue. Conclusion These studies were vital to the next step of testing editing reagents in the fetal NHP with a goal of treating monogenic diseases before birth.
Collapse
Affiliation(s)
- David A. Eaton
- Departments of Surgery, Yale School of Medicine, New Haven, CT, USA
- Departments of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Anna Y. Lynn
- Departments of Surgery, Yale School of Medicine, New Haven, CT, USA
| | | | - Emily I. Deschenes
- Departments of Surgery, Yale School of Medicine, New Haven, CT, USA
- Departments of Biomedical Engineering, Yale University, New Haven, CT, USA
| | | | - Rachel Rivero
- Departments of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Nicholas K. Yung
- Departments of Surgery, Yale School of Medicine, New Haven, CT, USA
| | | | - Peter M. Glazer
- Departments of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
- Departments of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Mert Ozan Bahtiyar
- Departments of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - W. Mark Saltzman
- Departments of Biomedical Engineering, Yale University, New Haven, CT, USA
- Departments of Chemical and Environmental Engineering, Yale University, New Haven, CT, USA
- Departments of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
- Departments of Dermatology, Yale University, New Haven, CT, USA
| | - David H. Stitelman
- Departments of Surgery, Yale School of Medicine, New Haven, CT, USA
- Departments of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
6
|
Walkey CJ. Fluorescent reporter mouse models for genome editing: choices and challenges. Mamm Genome 2024:10.1007/s00335-024-10079-8. [PMID: 39467961 DOI: 10.1007/s00335-024-10079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Affiliation(s)
- Christopher J Walkey
- Baylor College of Medicine, Department of Integrative Physiology, Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Chen K, Han H, Zhao S, Xu B, Yin B, Lawanprasert A, Trinidad M, Burgstone BW, Murthy N, Doudna JA. Lung and liver editing by lipid nanoparticle delivery of a stable CRISPR-Cas9 ribonucleoprotein. Nat Biotechnol 2024:10.1038/s41587-024-02437-3. [PMID: 39415058 PMCID: PMC12000389 DOI: 10.1038/s41587-024-02437-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 09/18/2024] [Indexed: 10/18/2024]
Abstract
Lipid nanoparticle (LNP) delivery of clustered regularly interspaced short palindromic repeat (CRISPR) ribonucleoproteins (RNPs) could enable high-efficiency, low-toxicity and scalable in vivo genome editing if efficacious RNP-LNP complexes can be reliably produced. Here we engineer a thermostable Cas9 from Geobacillus stearothermophilus (GeoCas9) to generate iGeoCas9 variants capable of >100× more genome editing of cells and organs compared with the native GeoCas9 enzyme. Furthermore, iGeoCas9 RNP-LNP complexes edit a variety of cell types and induce homology-directed repair in cells receiving codelivered single-stranded DNA templates. Using tissue-selective LNP formulations, we observe genome-editing levels of 16‒37% in the liver and lungs of reporter mice that receive single intravenous injections of iGeoCas9 RNP-LNPs. In addition, iGeoCas9 RNPs complexed to biodegradable LNPs edit the disease-causing SFTPC gene in lung tissue with 19% average efficiency, representing a major improvement over genome-editing levels observed previously using viral or nonviral delivery strategies. These results show that thermostable Cas9 RNP-LNP complexes can expand the therapeutic potential of genome editing.
Collapse
Affiliation(s)
- Kai Chen
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
| | - Hesong Han
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Sheng Zhao
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Bryant Xu
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
| | - Boyan Yin
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Atip Lawanprasert
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Marena Trinidad
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California Berkeley, Berkeley, CA, USA
| | - Benjamin W Burgstone
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Niren Murthy
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA.
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA.
| | - Jennifer A Doudna
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA.
- Innovative Genomics Institute, University of California Berkeley, Berkeley, CA, USA.
- Howard Hughes Medical Institute, University of California Berkeley, Berkeley, CA, USA.
- Gladstone Institutes, San Francisco, CA, USA.
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA.
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- Department of Chemistry, University of California Berkeley, Berkeley, CA, USA.
- California Institute for Quantitative Biosciences, University of California Berkeley, Berkeley, CA, USA.
| |
Collapse
|
8
|
Chaudhary N, Newby AN, Whitehead KA. Non-Viral RNA Delivery During Pregnancy: Opportunities and Challenges. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306134. [PMID: 38145340 PMCID: PMC11196389 DOI: 10.1002/smll.202306134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/25/2023] [Indexed: 12/26/2023]
Abstract
During pregnancy, the risk of maternal and fetal adversities increases due to physiological changes, genetic predispositions, environmental factors, and infections. Unfortunately, treatment options are severely limited because many essential interventions are unsafe, inaccessible, or lacking in sufficient scientific data to support their use. One potential solution to this challenge may lie in emerging RNA therapeutics for gene therapy, protein replacement, maternal vaccination, fetal gene editing, and other prenatal treatment applications. In this review, the current landscape of RNA platforms and non-viral RNA delivery technologies that are under active development for administration during pregnancy is explored. Advancements of pregnancy-specific RNA drugs against SARS-CoV-2, Zika, influenza, preeclampsia, and for in-utero gene editing are discussed. Finally, this study highlights bottlenecks that are impeding translation efforts of RNA therapies, including the lack of accurate cell-based and animal models of human pregnancy and concerns related to toxicity and immunogenicity during pregnancy. Overcoming these challenges will facilitate the rapid development of this new class of pregnancy-safe drugs.
Collapse
Affiliation(s)
- Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Alexandra N. Newby
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Kathryn A. Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213
| |
Collapse
|
9
|
Kong C, Yin G, Wang X, Sun Y. In Utero Gene Therapy and its Application in Genetic Hearing Loss. Adv Biol (Weinh) 2024; 8:e2400193. [PMID: 39007241 DOI: 10.1002/adbi.202400193] [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: 04/08/2024] [Revised: 07/03/2024] [Indexed: 07/16/2024]
Abstract
For monogenic genetic diseases, in utero gene therapy (IUGT) shows the potential for early prevention against irreversible and lethal pathological changes. Moreover, animal models have also demonstrated the effectiveness of IUGT in the treatment of coagulation disorders, hemoglobinopathies, neurogenetic disorders, and metabolic and pulmonary diseases. For major alpha thalassemia and severe osteogenesis imperfecta, in utero stem cell transplantation has entered the phase I clinical trial stage. Within the realm of the inner ear, genetic hearing loss significantly hampers speech, cognitive, and intellectual development in children. Nowadays, gene therapies offer substantial promise for deafness, with the success of clinical trials in autosomal recessive deafness 9 using AAV-OTOF gene therapy. However, the majority of genetic mutations that cause deafness affect the development of cochlear structures before the birth of fetuses. Thus, gene therapy before alterations in cochlear structure leading to hearing loss has promising applications. In this review, addressing advances in various fields of IUGT, the progress, and application of IUGT in the treatment of genetic hearing loss are focused, in particular its implementation methods and unique advantages.
Collapse
Affiliation(s)
- Chenyang Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ge Yin
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaohui Wang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Otorhinolaryngology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
10
|
Predella C, Lapsley L, Ni K, Murray JW, Liu HY, Motelow JE, Snoeck HW, Glasser SW, Saqi A, Dorrello NV. Engraftment of wild-type alveolar type II epithelial cells in surfactant protein C deficient mice. RESEARCH SQUARE 2024:rs.3.rs-4673915. [PMID: 39315275 PMCID: PMC11419168 DOI: 10.21203/rs.3.rs-4673915/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Childhood interstitial lung disease (chILD) secondary to pulmonary surfactant deficiency is a devastating chronic lung disease in children. Clinical presentation includes mild to severe respiratory failure and fibrosis. There is no specific treatment, except lung transplantation, which is hampered by a severe shortage of donor organs, especially for young patients. Repair of lungs with chILD represents a longstanding therapeutic challenge but cell therapy is a promising strategy. As surfactant is produced by alveolar type II epithelial (ATII) cells, engraftment with normal or gene-corrected ATII cells might provide an avenue to cure. Here we used a chILD disease-like model, Sftpc -/- mice, to provide proof-of-principle for this approach. Sftpc -/- mice developed chronic interstitial lung disease with age and were hypersensitive to bleomycin. We could engraft wild-type ATII cells after low dose bleomycin conditioning. Transplanted ATII cells produced mature SPC and attenuated bleomycin-induced lung injury up to two months post-transplant. This study demonstrates that partial replacement of mutant ATII cells can promote lung repair in a mouse model of chILD-like disease.
Collapse
Affiliation(s)
- Camilla Predella
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Chemistry, Materials and Chemical Engineering “G. Natta”, Politecnico of Milan, Milan, Italy
- Department of Electronics, Information and Bioengineering, Politecnico of Milan, Milan, Italy
| | - Lauren Lapsley
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Keyue Ni
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - John W. Murray
- Columbia Center for Human Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Hsiao-Yun Liu
- Columbia Center for Human Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Joshua E. Motelow
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Morgan Stanley Children’s Hospital, New York, NY, USA
| | - Hans-Willem Snoeck
- Columbia Center for Human Development, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Stephan W. Glasser
- Medical Sciences Program, Department of Medical Education, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Anjali Saqi
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian, New York, NY, USA
| | - N. Valerio Dorrello
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Division of Pediatric Critical Care Medicine and Hospital Medicine, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons and New York-Presbyterian Morgan Stanley Children’s Hospital, New York, NY, USA
| |
Collapse
|
11
|
Li S, Brakebusch C. Reporter Mice for Gene Editing: A Key Tool for Advancing Gene Therapy of Rare Diseases. Cells 2024; 13:1508. [PMID: 39273078 PMCID: PMC11394079 DOI: 10.3390/cells13171508] [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: 07/17/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
Most rare diseases are caused by mutations and can have devastating consequences. Precise gene editing by CRISPR/Cas is an exciting possibility for helping these patients, if no irreversible developmental defects have occurred. To optimize gene editing therapy, reporter mice for gene editing have been generated which, by expression of reporter genes, indicate the efficiency of precise and imprecise gene editing. These mice are important tools for testing and comparing novel gene editing methodologies. This review provides a comprehensive overview of reporter mice for gene editing which all have been used for monitoring CRISPR/Cas-mediated gene editing involving DNA double-strand breaks (DSBs). Furthermore, we discuss how reporter mice can be used for quickly checking genetic alterations by base editing (BE) or prime editing (PE).
Collapse
Affiliation(s)
| | - Cord Brakebusch
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen, Denmark;
| |
Collapse
|
12
|
Li R, Sone N, Gotoh S, Sun X, Hagood JS. Contemporary and emerging technologies for research in children's rare and interstitial lung disease. Pediatr Pulmonol 2024; 59:2349-2359. [PMID: 37204232 DOI: 10.1002/ppul.26490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/20/2023]
Abstract
Although recent decades have seen the identification, classification and discovery of the genetic basis of many children's interstitial and rare lung disease (chILD) disorders, detailed understanding of pathogenesis and specific therapies are still lacking for most of them. Fortunately, a revolution of technological advancements has created new opportunities to address these critical knowledge gaps. High-throughput sequencing has facilitated analysis of transcription of thousands of genes in thousands of single cells, creating tremendous breakthroughs in understanding normal and diseased cellular biology. Spatial techniques allow analysis of transcriptomes and proteomes at the subcellular level in the context of tissue architecture, in many cases even in formalin-fixed, paraffin-embedded specimens. Gene editing techniques allow creation of "humanized" animal models in a shorter time frame, for improved knowledge and preclinical therapeutic testing. Regenerative medicine approaches and bioengineering advancements facilitate the creation of patient-derived induced pluripotent stem cells and their differentiation into tissue-specific cell types which can be studied in multicellular "organoids" or "organ-on-a-chip" approaches. These technologies, singly and in combination, are already being applied to gain new biological insights into chILD disorders. The time is ripe to systematically apply these technologies to chILD, together with sophisticated data science approaches, to improve both biological understanding and disease-specific therapy.
Collapse
Affiliation(s)
- Rongbo Li
- Department of Pediatrics, Division of Respiratory Medicine, UC-San Diego, La Jolla, California, USA
| | - Naoyuki Sone
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Shimpei Gotoh
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Xin Sun
- Department of Pediatrics, Division of Respiratory Medicine, UC-San Diego, La Jolla, California, USA
| | - James S Hagood
- Department of Pediatrics, Pulmonology Division, Program for Rare and Interstitial Lung Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
13
|
Wambach JA, Vece TJ. Clinical and research innovations in childhood interstitial lung disease (chILD). Pediatr Pulmonol 2024; 59:2233-2235. [PMID: 38651871 PMCID: PMC11324416 DOI: 10.1002/ppul.27025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Affiliation(s)
- Jennifer A Wambach
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, Saint Louis, Missouri, USA
| | - Timothy J Vece
- Department of Pediatrics, University of North Carolina-Chapel Hill, Chapel Hill, USA
| |
Collapse
|
14
|
Ajaykumar CB, Rajkumar S, Suresh B, Birappa G, Gowda DAA, Jayachandran A, Kim KS, Hong SH, Ramakrishna S. Advances in applications of the CRISPR/Cas9 system for respiratory diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 210:127-147. [PMID: 39824578 DOI: 10.1016/bs.pmbts.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2025]
Abstract
Genetic and environmental factors can have an impact on lung and respiratory disorders which are associated with severe symptoms and have high mortality rates. Many respiratory diseases are significantly influenced by genetic or epigenetic factors. Gene therapy offers a powerful approach providing therapeutic treatment for lung diseases. Clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (CRISPR/Cas9) are promising gene modifying tool that can edit the genome. The utilization of CRISPR/Cas9 systems in the investigation of respiratory disorders has resulted in advancements such as the rectification of deleterious mutations in patient-derived cells and the alteration of genes in multiple mammalian lung disease models. New avenues of treatment for lung disorders have been opened up by advances in CRISPR/Cas9 research. In this chapter, we discuss the known genes and mutations that cause several common respiratory disorders such as COPD, asthma, IPF, and ARDS. We further review the current research using CRISPR/Cas9 in numerous respiratory disorders and possible therapeutic treatments.
Collapse
Affiliation(s)
- C Bindu Ajaykumar
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Sripriya Rajkumar
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Bharathi Suresh
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Girish Birappa
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - D A Ayush Gowda
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Aparna Jayachandran
- Fiona Elsey Cancer Research Institute, VIC, Australia; Federation University, VIC, Australia
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; College of Medicine, Hanyang University, Seoul, Korea.
| | | | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; College of Medicine, Hanyang University, Seoul, Korea.
| |
Collapse
|
15
|
Palanki R, Riley JS, Bose SK, Luks V, Dave A, Kus N, White BM, Ricciardi AS, Swingle KL, Xue L, Sung D, Thatte AS, Safford HC, Chaluvadi VS, Carpenter M, Han EL, Maganti R, Hamilton AG, Mrksich K, Billingsley MB, Zoltick PW, Alameh MG, Weissman D, Mitchell MJ, Peranteau WH. In utero delivery of targeted ionizable lipid nanoparticles facilitates in vivo gene editing of hematopoietic stem cells. Proc Natl Acad Sci U S A 2024; 121:e2400783121. [PMID: 39078677 PMCID: PMC11317576 DOI: 10.1073/pnas.2400783121] [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: 02/11/2024] [Accepted: 06/28/2024] [Indexed: 07/31/2024] Open
Abstract
Monogenic blood diseases are among the most common genetic disorders worldwide. These diseases result in significant pediatric and adult morbidity, and some can result in death prior to birth. Novel ex vivo hematopoietic stem cell (HSC) gene editing therapies hold tremendous promise to alter the therapeutic landscape but are not without potential limitations. In vivo gene editing therapies offer a potentially safer and more accessible treatment for these diseases but are hindered by a lack of delivery vectors targeting HSCs, which reside in the difficult-to-access bone marrow niche. Here, we propose that this biological barrier can be overcome by taking advantage of HSC residence in the easily accessible liver during fetal development. To facilitate the delivery of gene editing cargo to fetal HSCs, we developed an ionizable lipid nanoparticle (LNP) platform targeting the CD45 receptor on the surface of HSCs. After validating that targeted LNPs improved messenger ribonucleic acid (mRNA) delivery to hematopoietic lineage cells via a CD45-specific mechanism in vitro, we demonstrated that this platform mediated safe, potent, and long-term gene modulation of HSCs in vivo in multiple mouse models. We further optimized this LNP platform in vitro to encapsulate and deliver CRISPR-based nucleic acid cargos. Finally, we showed that optimized and targeted LNPs enhanced gene editing at a proof-of-concept locus in fetal HSCs after a single in utero intravenous injection. By targeting HSCs in vivo during fetal development, our Systematically optimized Targeted Editing Machinery (STEM) LNPs may provide a translatable strategy to treat monogenic blood diseases before birth.
Collapse
Affiliation(s)
- Rohan Palanki
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - John S. Riley
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Sourav K. Bose
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Valerie Luks
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Apeksha Dave
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Nicole Kus
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Brandon M. White
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Adele S. Ricciardi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Kelsey L. Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Derek Sung
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Ajay S. Thatte
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Hannah C. Safford
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Venkata S. Chaluvadi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Marco Carpenter
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Emily L. Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Rohin Maganti
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Alex G. Hamilton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - Kaitlin Mrksich
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | | | - Philip W. Zoltick
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Mohamad-Gabriel Alameh
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
| | - William H. Peranteau
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA19104
| |
Collapse
|
16
|
Lara-Sáez I, Mencía Á, Recuero E, Li Y, García M, Oteo M, Gallego MI, Enguita AB, de Prado-Verdún D, A S, Wang W, García-Escudero R, Murillas R, Santos M. Nonviral CRISPR/Cas9 mutagenesis for streamlined generation of mouse lung cancer models. Proc Natl Acad Sci U S A 2024; 121:e2322917121. [PMID: 38959035 PMCID: PMC11252735 DOI: 10.1073/pnas.2322917121] [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: 01/09/2024] [Accepted: 06/14/2024] [Indexed: 07/04/2024] Open
Abstract
Functional analysis in mouse models is necessary to establish the involvement of a set of genetic variations in tumor development. A modeling platform to facilitate and cost-effectively analyze the role of multiple genes in carcinogenesis would be valuable. Here, we present an innovative strategy for lung mutagenesis using CRISPR/Cas9 ribonucleoproteins delivered via cationic polymers. This approach allows the simultaneous inactivation of multiple genes. We validate the effectiveness of this system by targeting a group of tumor suppressor genes, specifically Rb1, Rbl1, Pten, and Trp53, which were chosen for their potential to cause lung tumors, namely small cell lung carcinoma (SCLC). Tumors with histologic and transcriptomic features of human SCLC emerged after intratracheal administration of CRISPR/polymer nanoparticles. These tumors carried loss-of-function mutations in all four tumor suppressor genes at the targeted positions. These findings were reproduced in two different pure genetic backgrounds. We provide a proof of principle for simplified modeling of lung tumorigenesis to facilitate functional testing of potential cancer-related genes.
Collapse
Affiliation(s)
- Irene Lara-Sáez
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, DublinD04 V1W8, Ireland
| | - Ángeles Mencía
- Biomedical Innovation Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid28040, Spain
- CB06/07/0019 Unit, Centro de Investigación Biomédica en Red en Enfermedades Raras, Madrid28029, Spain
- Regenerative Medicine and Tissue Bioengineering Group, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid28040, Spain
| | - Enrique Recuero
- Biomedical Innovation Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid28040, Spain
- Cellular and Molecular Genitourinary Oncology Group, Institute of Biomedical Research Hospital “12 de Octubre”, Madrid28041, Spain
| | - Yinghao Li
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, DublinD04 V1W8, Ireland
| | - Marta García
- CB06/07/0019 Unit, Centro de Investigación Biomédica en Red en Enfermedades Raras, Madrid28029, Spain
- Regenerative Medicine and Tissue Bioengineering Group, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid28040, Spain
- Department of Biomedical Engineering, Polytechnic School, Carlos III University, Leganés, Madrid28911, Spain
| | - Marta Oteo
- Biomedical Applications and Pharmacokinetics Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid28040, Spain
| | - Marta I. Gallego
- Unidad de Histología, Unidades Centrales Científico Tecnológicas, Instituto de Salud Carlos III, Madrid28220, Spain
| | - Ana Belén Enguita
- Pathology Department, University Hospital “12 de Octubre”, Madrid28041, Spain
| | - Diana de Prado-Verdún
- Biomedical Innovation Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid28040, Spain
- CB06/07/0019 Unit, Centro de Investigación Biomédica en Red en Enfermedades Raras, Madrid28029, Spain
- Regenerative Medicine and Tissue Bioengineering Group, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid28040, Spain
| | - Sigen A
- Research and Clinical Translation Center of Gene Medicine and Tissue Engineering, School of Public Health, Anhui University of Science and Technology, Huainan232001, China
| | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, DublinD04 V1W8, Ireland
| | - Ramón García-Escudero
- Biomedical Innovation Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid28040, Spain
- Cellular and Molecular Genitourinary Oncology Group, Institute of Biomedical Research Hospital “12 de Octubre”, Madrid28041, Spain
- Tumor Progression Mechanisms Program, Centro de Investigación Biomédica en Red de Cáncer, Madrid28029, Spain
| | - Rodolfo Murillas
- Biomedical Innovation Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid28040, Spain
- CB06/07/0019 Unit, Centro de Investigación Biomédica en Red en Enfermedades Raras, Madrid28029, Spain
- Regenerative Medicine and Tissue Bioengineering Group, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid28040, Spain
| | - Mirentxu Santos
- Biomedical Innovation Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas, Madrid28040, Spain
- Cellular and Molecular Genitourinary Oncology Group, Institute of Biomedical Research Hospital “12 de Octubre”, Madrid28041, Spain
- Tumor Progression Mechanisms Program, Centro de Investigación Biomédica en Red de Cáncer, Madrid28029, Spain
| |
Collapse
|
17
|
Mattar CN, Chew WL, Lai PS. Embryo and fetal gene editing: Technical challenges and progress toward clinical applications. Mol Ther Methods Clin Dev 2024; 32:101229. [PMID: 38533521 PMCID: PMC10963250 DOI: 10.1016/j.omtm.2024.101229] [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] [Indexed: 03/28/2024]
Abstract
Gene modification therapies (GMTs) are slowly but steadily making progress toward clinical application. As the majority of rare diseases have an identified genetic cause, and as rare diseases collectively affect 5% of the global population, it is increasingly important to devise gene correction strategies to address the root causes of the most devastating of these diseases and to provide access to these novel therapies to the most affected populations. The main barriers to providing greater access to GMTs continue to be the prohibitive cost of developing these novel drugs at clinically relevant doses, subtherapeutic effects, and toxicity related to the specific agents or high doses required. In vivo strategy and treating younger patients at an earlier course of their disease could lower these barriers. Although currently regarded as niche specialties, prenatal and preconception GMTs offer a robust solution to some of these barriers. Indeed, treating either the fetus or embryo benefits from economy of scale, targeting pre-pathological tissues in the fetus prior to full pathogenesis, or increasing the likelihood of complete tissue targeting by correcting pluripotent embryonic cells. Here, we review advances in embryo and fetal GMTs and discuss requirements for clinical application.
Collapse
Affiliation(s)
- Citra N.Z. Mattar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, Singapore 119228
- Department of Obstetrics and Gynaecology, National University Health System, Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, Singapore 119228
| | - Wei Leong Chew
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, 60 Biopolis St, Singapore, Singapore 138672
| | - Poh San Lai
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, Singapore 119228
| |
Collapse
|
18
|
Sánchez Rivera FJ, Dow LE. How CRISPR Is Revolutionizing the Generation of New Models for Cancer Research. Cold Spring Harb Perspect Med 2024; 14:a041384. [PMID: 37487630 PMCID: PMC11065179 DOI: 10.1101/cshperspect.a041384] [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: 07/26/2023]
Abstract
Cancers arise through acquisition of mutations in genes that regulate core biological processes like cell proliferation and cell death. Decades of cancer research have led to the identification of genes and mutations causally involved in disease development and evolution, yet defining their precise function across different cancer types and how they influence therapy responses has been challenging. Mouse models have helped define the in vivo function of cancer-associated alterations, and genome-editing approaches using CRISPR have dramatically accelerated the pace at which these models are developed and studied. Here, we highlight how CRISPR technologies have impacted the development and use of mouse models for cancer research and discuss the many ways in which these rapidly evolving platforms will continue to transform our understanding of this disease.
Collapse
Affiliation(s)
- Francisco J Sánchez Rivera
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Lukas E Dow
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10065, USA
- Department of Biochemistry, Weill Cornell Medicine, New York, New York 10065, USA
- Department of Medicine, Weill Cornell Medicine, New York, New York 10065, USA
| |
Collapse
|
19
|
Basil MC, Alysandratos KD, Kotton DN, Morrisey EE. Lung repair and regeneration: Advanced models and insights into human disease. Cell Stem Cell 2024; 31:439-454. [PMID: 38492572 PMCID: PMC11070171 DOI: 10.1016/j.stem.2024.02.009] [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: 12/05/2023] [Revised: 02/07/2024] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
The respiratory system acts as both the primary site of gas exchange and an important sensor and barrier to the external environment. The increase in incidences of respiratory disease over the past decades has highlighted the importance of developing improved therapeutic approaches. This review will summarize recent research on the cellular complexity of the mammalian respiratory system with a focus on gas exchange and immunological defense functions of the lung. Different models of repair and regeneration will be discussed to help interpret human and animal data and spur the investigation of models and assays for future drug development.
Collapse
Affiliation(s)
- Maria C Basil
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn, Children's Hospital of Philadelphia (CHOP) Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Konstantinos-Dionysios Alysandratos
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA.
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA.
| | - Edward E Morrisey
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn, Children's Hospital of Philadelphia (CHOP) Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Sun YL, Hennessey EE, Heins H, Yang P, Villacorta-Martin C, Kwan J, Gopalan K, James M, Emili A, Cole FS, Wambach JA, Kotton DN. Human pluripotent stem cell modeling of alveolar type 2 cell dysfunction caused by ABCA3 mutations. J Clin Invest 2024; 134:e164274. [PMID: 38226623 PMCID: PMC10786693 DOI: 10.1172/jci164274] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/14/2023] [Indexed: 01/17/2024] Open
Abstract
Mutations in ATP-binding cassette A3 (ABCA3), a phospholipid transporter critical for surfactant homeostasis in pulmonary alveolar type II epithelial cells (AEC2s), are the most common genetic causes of childhood interstitial lung disease (chILD). Treatments for patients with pathological variants of ABCA3 mutations are limited, in part due to a lack of understanding of disease pathogenesis resulting from an inability to access primary AEC2s from affected children. Here, we report the generation of AEC2s from affected patient induced pluripotent stem cells (iPSCs) carrying homozygous versions of multiple ABCA3 mutations. We generated syngeneic CRISPR/Cas9 gene-corrected and uncorrected iPSCs and ABCA3-mutant knockin ABCA3:GFP fusion reporter lines for in vitro disease modeling. We observed an expected decreased capacity for surfactant secretion in ABCA3-mutant iPSC-derived AEC2s (iAEC2s), but we also found an unexpected epithelial-intrinsic aberrant phenotype in mutant iAEC2s, presenting as diminished progenitor potential, increased NFκB signaling, and the production of pro-inflammatory cytokines. The ABCA3:GFP fusion reporter permitted mutant-specific, quantifiable characterization of lamellar body size and ABCA3 protein trafficking, functional features that are perturbed depending on ABCA3 mutation type. Our disease model provides a platform for understanding ABCA3 mutation-mediated mechanisms of alveolar epithelial cell dysfunction that may trigger chILD pathogenesis.
Collapse
Affiliation(s)
- Yuliang L. Sun
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Erin E. Hennessey
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Hillary Heins
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine and St. Louis Children’s Hospital, St. Louis, Missouri, USA
| | - Ping Yang
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine and St. Louis Children’s Hospital, St. Louis, Missouri, USA
| | - Carlos Villacorta-Martin
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - Julian Kwan
- Departments of Biology and Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Krithi Gopalan
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Marianne James
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - Andrew Emili
- Departments of Biology and Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - F. Sessions Cole
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine and St. Louis Children’s Hospital, St. Louis, Missouri, USA
| | - Jennifer A. Wambach
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine and St. Louis Children’s Hospital, St. Louis, Missouri, USA
| | - Darrell N. Kotton
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Chen Z, Kwan SY, Mir A, Hazeltine M, Shin M, Liang SQ, Chan IL, Kelly K, Ghanta KS, Gaston N, Cao Y, Xie J, Gao G, Xue W, Sontheimer EJ, Watts JK. A Fluorescent Reporter Mouse for In Vivo Assessment of Genome Editing with Diverse Cas Nucleases and Prime Editors. CRISPR J 2023; 6:570-582. [PMID: 38108517 PMCID: PMC10753986 DOI: 10.1089/crispr.2023.0048] [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: 07/27/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
CRISPR-based genome-editing technologies, including nuclease editing, base editing, and prime editing, have recently revolutionized the development of therapeutics targeting disease-causing mutations. To advance the assessment and development of genome editing tools, a robust mouse model is valuable, particularly for evaluating in vivo activity and delivery strategies. In this study, we successfully generated a knock-in mouse line carrying the Traffic Light Reporter design known as TLR-multi-Cas variant 1 (TLR-MCV1). We comprehensively validated the functionality of this mouse model for both in vitro and in vivo nuclease and prime editing. The TLR-MCV1 reporter mouse represents a versatile and powerful tool for expediting the development of editing technologies and their therapeutic applications.
Collapse
Affiliation(s)
- Zexiang Chen
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Suet-Yan Kwan
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Aamir Mir
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Max Hazeltine
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Minwook Shin
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Shun-Qing Liang
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Io Long Chan
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Karen Kelly
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Krishna S. Ghanta
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Nicholas Gaston
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Yueying Cao
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Jun Xie
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Guangping Gao
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Wen Xue
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Erik J. Sontheimer
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Jonathan K. Watts
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
22
|
Peers de Nieuwburgh M, Wambach JA, Griese M, Danhaive O. Towards personalized therapies for genetic disorders of surfactant dysfunction. Semin Fetal Neonatal Med 2023; 28:101500. [PMID: 38036307 PMCID: PMC10753445 DOI: 10.1016/j.siny.2023.101500] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Genetic disorders of surfactant dysfunction are a rare cause of chronic, progressive or refractory respiratory failure in term and preterm infants. This review explores genetic mechanisms underpinning surfactant dysfunction, highlighting specific surfactant-associated genes including SFTPB, SFTPC, ABCA3, and NKX2.1. Pathogenic variants in these genes contribute to a range of clinical presentations and courses, from neonatal hypoxemic respiratory failure to childhood interstitial lung disease and even adult-onset pulmonary fibrosis. This review emphasizes the importance of early recognition, thorough phenotype assessment, and assessment of variant functionality as essential prerequisites for treatments including lung transplantation. We explore emerging treatment options, including personalized pharmacological approaches and gene therapy strategies. In conclusion, this comprehensive review offers valuable insights into the pathogenic mechanisms of genetic disorders of surfactant dysfunction, genetic fundamentals, available and emerging therapeutic options, and underscores the need for further research to develop personalized therapies for affected infants and children.
Collapse
Affiliation(s)
- Maureen Peers de Nieuwburgh
- Division of Neonatology, Department of Pediatrics, St-Luc University Hospital, Catholic University of Louvain, Brussels, Belgium.
| | - Jennifer A Wambach
- Washington University School of Medicine/St. Louis Children's Hospital, One Children's Place, St. Louis, Missouri, USA.
| | - Matthias Griese
- Pediatric Pulmonology, Dr von Hauner Children's Hospital, University-Hospital, German Center for Lung Research (DZL), Munich, Germany.
| | - Olivier Danhaive
- Division of Neonatology, Department of Pediatrics, St-Luc University Hospital, Catholic University of Louvain, Brussels, Belgium; Division of Neonatology, Benioff Children's Hospital, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
23
|
Gogoleva N, Shahri ZJ, Noda A, Liao CW, Wakimoto A, Inoue Y, Jeon H, Takahashi S, Hamada M. Intraplacental injection of AAV9-CMV-iCre results in the widespread transduction of multiple organs in double-reporter mouse embryos. Exp Anim 2023; 72:460-467. [PMID: 37183025 PMCID: PMC10658086 DOI: 10.1538/expanim.23-0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/29/2023] [Indexed: 05/16/2023] Open
Abstract
Adeno-associated virus serotype 9 (AAV9) has become a popular tool for gene transfer because of its ability to cross the blood-brain barrier and efficiently transduce genetic material into a variety of cell types. The study utilized GRR (Green-to-Red Reporter) mouse embryos, in which the expression of iCre results in the disappearance of Green Fluorescent Protein (GFP) expression and the detection of Discosoma sp. Red Fluorescent Protein (DsRed) expression by intraplacental injection. Our results demonstrate that AAV9-CMV-iCre can transduce multiple organs in embryos at developmental stages E9.5-E11.5, including the liver, heart, brain, thymus, and intestine. These findings suggest that intraplacental injection of AAV9-CMV-iCre is a viable method for the widespread transduction of GRR mouse embryos.
Collapse
Affiliation(s)
- Natalia Gogoleva
- Ph.D. Program in Human Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Zeynab Javanfekr Shahri
- Ph.D. Program in Human Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Atsushi Noda
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Ching-Wei Liao
- Ph.D. Program in Human Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Arata Wakimoto
- Ph.D. Program in Human Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuri Inoue
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hyojung Jeon
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Laboratory of Stem Cell Therapy, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Laboratory Animal Resource Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Michito Hamada
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Laboratory Animal Resource Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
24
|
Musunuru K. Playing the genetic lottery: an interview with Kiran Musunuru. Dis Model Mech 2023; 16:dmm050508. [PMID: 37814839 PMCID: PMC10581381 DOI: 10.1242/dmm.050508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023] Open
Affiliation(s)
- Kiran Musunuru
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
25
|
Konkimalla A, Elmore Z, Konishi S, Macadlo L, Katsura H, Tata A, Asokan A, Tata PR. Efficient Adeno-associated Virus-mediated Transgenesis in Alveolar Stem Cells and Associated Niches. Am J Respir Cell Mol Biol 2023; 69:255-265. [PMID: 37315312 PMCID: PMC10503306 DOI: 10.1165/rcmb.2022-0424ma] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 06/13/2023] [Indexed: 06/16/2023] Open
Abstract
Targeted delivery of transgenes to tissue-resident stem cells and related niches offers avenues for interrogating pathways and editing endogenous alleles for therapeutic interventions. Here, we survey multiple adeno-associated virus (AAV) serotypes, administered via intranasal and retroorbital routes in mice, to target lung alveolar stem cell niches. We found that AAV5, AAV4, and AAV8 efficiently and preferentially transduce alveolar type-2 stem cells (AT2s), endothelial cells, and PDGFRA+ fibroblasts, respectively. Notably, some AAVs show different cell tropisms depending on the route of administration. Proof-of-concept experiments reveal the versatility of AAV5-mediated transgenesis for AT2-lineage labeling, clonal cell tracing after cell ablation, and conditional gene inactivation in both postnatal and adult mouse lungs in vivo. AAV6, but not AAV5, efficiently transduces both mouse and human AT2s in alveolar organoid cultures. Furthermore, AAV5 and AAV6 can be used to deliver guide RNAs and transgene cassettes for homologous recombination in vivo and ex vivo, respectively. Using this system coupled with clonal derivation of AT2 organoids, we demonstrate efficient and simultaneous editing of multiple loci, including targeted insertion of a payload cassette in AT2s. Taken together, our studies highlight the powerful utility of AAVs for interrogating alveolar stem cells and other specific cell types both in vivo and ex vivo.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Aravind Asokan
- Department of Surgery
- Department of Molecular Genetics and Microbiology
- Department of Biomedical Engineering
- Center for Advanced Genomic Technologies, and
- Duke Regeneration Center, Duke University, Durham, North Carolina
| | - Purushothama Rao Tata
- Department of Cell Biology
- Duke Cancer Institute, and
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- Center for Advanced Genomic Technologies, and
- Duke Regeneration Center, Duke University, Durham, North Carolina
| |
Collapse
|
26
|
Zhang Y, Peng Q, Zhang R, Li C, Xu Q, Xia L, Wang Y, Liu P, Pan H. Advances in CRISPR/Cas-Based Strategies on Zoonosis. Transbound Emerg Dis 2023; 2023:9098445. [PMID: 40303816 PMCID: PMC12017210 DOI: 10.1155/2023/9098445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 05/04/2023] [Accepted: 07/05/2023] [Indexed: 05/02/2025]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) has emerged as the predominant technique for gene editing technique due to its high efficiency and low cost. In the area of zoonosis, CRISPR/Cas is also widely used in different research areas. This paper reviewed the principles of CRISPR/Cas technique and its applications in zoonosis. Moreover, we analyze the shortcomings and weaknesses that currently limit its use, highlight its direction for improvement, and foresee its application prospects in the prevention and treatment of zoonosis. For the purpose of preventing and controlling zoonosis, we need to develop diagnostic method with high sensitivity and specificity, highly protective vaccines, and also better understanding of the pathogenesis. Our review aimed to promote the application and improvement of CRISPR/Cas technique in the above-mentioned areas, and provide brief and comprehensive references for CRISPR/Cas-based research. Through reviewing the advances in CRISPR/Cas-based strategies on zoonosis, we believe that CRISPR/Cas technique will provide more powerful assistance for the prevention and control of zoonosis.
Collapse
Affiliation(s)
- Yi Zhang
- Institute of Zoonosis, College of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi, China
| | - Qifeng Peng
- Institute of Zoonosis, College of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi, China
| | - Renjun Zhang
- Center for Animal Disease Control and Prevention of Guizhou Province, Guiyang, China
| | - Chao Li
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Quangang Xu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Luming Xia
- Center for Animal Disease Control and Prevention of Shanghai City, Shanghai, China
| | - Youming Wang
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Ping Liu
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Hong Pan
- Institute of Zoonosis, College of Public Health, Zunyi Medical University, Zunyi, China
- Key Laboratory of Maternal and Child Health and Exposure Science of Guizhou Higher Education Institutes, Zunyi, China
| |
Collapse
|
27
|
Palanki R, Bose SK, Dave A, White BM, Berkowitz C, Luks V, Yaqoob F, Han E, Swingle KL, Menon P, Hodgson E, Biswas A, Billingsley MM, Li L, Yiping F, Carpenter M, Trokhan A, Yeo J, Johana N, Wan TY, Alameh MG, Bennett FC, Storm PB, Jain R, Chan J, Weissman D, Mitchell MJ, Peranteau WH. Ionizable Lipid Nanoparticles for Therapeutic Base Editing of Congenital Brain Disease. ACS NANO 2023; 17:13594-13610. [PMID: 37458484 PMCID: PMC11025390 DOI: 10.1021/acsnano.3c02268] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Delivery of mRNA-based therapeutics to the perinatal brain holds great potential in treating congenital brain diseases. However, nonviral delivery platforms that facilitate nucleic acid delivery in this environment have yet to be rigorously studied. Here, we screen a diverse library of ionizable lipid nanoparticles (LNPs) via intracerebroventricular (ICV) injection in both fetal and neonatal mice and identify an LNP formulation with greater functional mRNA delivery in the perinatal brain than an FDA-approved industry standard LNP. Following in vitro optimization of the top-performing LNP (C3 LNP) for codelivery of an adenine base editing platform, we improve the biochemical phenotype of a lysosomal storage disease in the neonatal mouse brain, exhibit proof-of-principle mRNA brain transfection in vivo in a fetal nonhuman primate model, and demonstrate the translational potential of C3 LNPs ex vivo in human patient-derived brain tissues. These LNPs may provide a clinically translatable platform for in utero and postnatal mRNA therapies including gene editing in the brain.
Collapse
Affiliation(s)
- Rohan Palanki
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sourav K Bose
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Apeksha Dave
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brandon M. White
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Cara Berkowitz
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Valerie Luks
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Fazeela Yaqoob
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emily Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kelsey L Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pallavi Menon
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Emily Hodgson
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Arijit Biswas
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
| | | | - Li Li
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fan Yiping
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
| | - Marco Carpenter
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexandra Trokhan
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Julie Yeo
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
| | | | - Tan Yi Wan
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
| | - Mohamad-Gabriel Alameh
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Frederick Chris Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Phillip B. Storm
- Division of Neurosurgery, Children’s Hospital of Philadelphia, PA 19104, USA
| | - Rajan Jain
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jerry Chan
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
- Department of Reproductive Medicine, KK Women’s and Children’s Hospital, Singapore, 229899, SG
| | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - William H. Peranteau
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, PA, USA
| |
Collapse
|
28
|
Stampone E, Bencivenga D, Capellupo MC, Roberti D, Tartaglione I, Perrotta S, Della Ragione F, Borriello A. Genome editing and cancer therapy: handling the hypoxia-responsive pathway as a promising strategy. Cell Mol Life Sci 2023; 80:220. [PMID: 37477829 PMCID: PMC10361942 DOI: 10.1007/s00018-023-04852-2] [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: 05/02/2023] [Revised: 06/14/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023]
Abstract
The precise characterization of oxygen-sensing pathways and the identification of pO2-regulated gene expression are both issues of critical importance. The O2-sensing system plays crucial roles in almost all the pivotal human processes, including the stem cell specification, the growth and development of tissues (such as embryogenesis), the modulation of intermediate metabolism (including the shift of the glucose metabolism from oxidative to anaerobic ATP production and vice versa), and the control of blood pressure. The solid cancer microenvironment is characterized by low oxygen levels and by the consequent activation of the hypoxia response that, in turn, allows a complex adaptive response characterized mainly by neoangiogenesis and metabolic reprogramming. Recently, incredible advances in molecular genetic methodologies allowed the genome editing with high efficiency and, above all, the precise identification of target cells/tissues. These new possibilities and the knowledge of the mechanisms of adaptation to hypoxia suggest the effective development of new therapeutic approaches based on the manipulation, targeting, and exploitation of the oxygen-sensor system molecular mechanisms.
Collapse
Affiliation(s)
- Emanuela Stampone
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 7, 80138, Naples, Italy
| | - Debora Bencivenga
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 7, 80138, Naples, Italy
| | - Maria Chiara Capellupo
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 7, 80138, Naples, Italy
| | - Domenico Roberti
- Department of the Woman, the Child and of the General and Specialty Surgery, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 2, 80138, Naples, Italy
| | - Immacolata Tartaglione
- Department of the Woman, the Child and of the General and Specialty Surgery, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 2, 80138, Naples, Italy
| | - Silverio Perrotta
- Department of the Woman, the Child and of the General and Specialty Surgery, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 2, 80138, Naples, Italy
| | - Fulvio Della Ragione
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 7, 80138, Naples, Italy.
| | - Adriana Borriello
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via Luigi De Crecchio, 7, 80138, Naples, Italy.
| |
Collapse
|
29
|
Gao K, Li J, Song H, Han H, Wang Y, Yin B, Farmer DL, Murthy N, Wang A. In utero delivery of mRNA to the heart, diaphragm and muscle with lipid nanoparticles. Bioact Mater 2023; 25:387-398. [PMID: 36844366 PMCID: PMC9950423 DOI: 10.1016/j.bioactmat.2023.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/26/2023] [Accepted: 02/11/2023] [Indexed: 02/19/2023] Open
Abstract
Nanoparticle-based drug delivery systems have the potential to revolutionize medicine, but their low vascular permeability and rapid clearance by phagocytic cells have limited their medical impact. Nanoparticles delivered at the in utero stage can overcome these key limitations due to the high rate of angiogenesis and cell division in fetal tissue and the under-developed immune system. However, very little is known about nanoparticle drug delivery at the fetal stage of development. In this report, using Ai9 CRE reporter mice, we demonstrate that lipid nanoparticle (LNP) mRNA complexes can deliver mRNA in utero, and can access and transfect major organs, such as the heart, the liver, kidneys, lungs and the gastrointestinal tract with remarkable efficiency and low toxicity. In addition, at 4 weeks after birth, we demonstrate that 50.99 ± 5.05%, 36.62 ± 3.42% and 23.7 ± 3.21% of myofiber in the diaphragm, heart and skeletal muscle, respectively, were transfected. Finally, we show here that Cas9 mRNA and sgRNA complexed to LNPs were able to edit the fetal organs in utero. These experiments demonstrate the possibility of non-viral delivery of mRNA to organs outside of the liver in utero, which provides a promising strategy for treating a wide variety of devastating diseases before birth.
Collapse
Affiliation(s)
- Kewa Gao
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Jie Li
- Department of Bioengineering, University of California, Berkeley, CA, 94704, United States
| | - Hengyue Song
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Hunan, 410013, China
| | - Hesong Han
- Department of Bioengineering, University of California, Berkeley, CA, 94704, United States
| | - Yongheng Wang
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, United States
| | - Boyan Yin
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Diana L. Farmer
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
| | - Niren Murthy
- Department of Bioengineering, University of California, Berkeley, CA, 94704, United States
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA, 95817, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, 95817, United States
- Department of Biomedical Engineering, University of California, Davis, CA, 95616, United States
| |
Collapse
|
30
|
Nakamura S, Inada E, Saitoh I, Sato M. Recent Genome-Editing Approaches toward Post-Implanted Fetuses in Mice. BIOTECH 2023; 12:biotech12020037. [PMID: 37218754 DOI: 10.3390/biotech12020037] [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: 03/06/2023] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 05/24/2023] Open
Abstract
Genome editing, as exemplified by the CRISPR/Cas9 system, has recently been employed to effectively generate genetically modified animals and cells for the purpose of gene function analysis and disease model creation. There are at least four ways to induce genome editing in individuals: the first is to perform genome editing at the early preimplantation stage, such as fertilized eggs (zygotes), for the creation of whole genetically modified animals; the second is at post-implanted stages, as exemplified by the mid-gestational stages (E9 to E15), for targeting specific cell populations through in utero injection of viral vectors carrying genome-editing components or that of nonviral vectors carrying genome-editing components and subsequent in utero electroporation; the third is at the mid-gestational stages, as exemplified by tail-vein injection of genome-editing components into the pregnant females through which the genome-editing components can be transmitted to fetal cells via a placenta-blood barrier; and the last is at the newborn or adult stage, as exemplified by facial or tail-vein injection of genome-editing components. Here, we focus on the second and third approaches and will review the latest techniques for various methods concerning gene editing in developing fetuses.
Collapse
Affiliation(s)
- Shingo Nakamura
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Saitama 359-8513, Japan
| | - Emi Inada
- Department of Pediatric Dentistry, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Issei Saitoh
- Department of Pediatric Dentistry, Asahi University School of Dentistry, Mizuho-shi 501-0296, Japan
| | - Masahiro Sato
- Department of Genome Medicine, National Center for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
31
|
Mattar CNZ, Chan JKY, Choolani M. Gene modification therapies for hereditary diseases in the fetus. Prenat Diagn 2023; 43:674-686. [PMID: 36965009 PMCID: PMC10946994 DOI: 10.1002/pd.6347] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/20/2023] [Accepted: 03/02/2023] [Indexed: 03/27/2023]
Abstract
Proof-of-principle disease models have demonstrated the feasibility of an intrauterine gene modification therapy (in utero gene therapy (IUGT)) approach to hereditary diseases as diverse as coagulation disorders, haemoglobinopathies, neurogenetic disorders, congenital metabolic, and pulmonary diseases. Gene addition, which requires the delivery of an integrating or episomal transgene to the target cell nucleus to be transcribed, and gene editing, where the mutation is corrected within the gene of origin, have both been used successfully to increase normal protein production in a bid to reverse or arrest pathology in utero. While most experimental models have employed lentiviral, adenoviral, and adeno-associated viral vectors engineered to efficiently enter target cells, newer models have also demonstrated the applicability of non-viral lipid nanoparticles. Amelioration of pathology is dependent primarily on achieving sustained therapeutic transgene expression, silencing of transgene expression, production of neutralising antibodies, the dilutional effect of the recipient's growth on the mass of transduced cells, and the degree of pre-existing cellular damage. Safety assessment of any IUGT strategy will require long-term postnatal surveillance of both the fetal recipient and the maternal bystander for cell and genome toxicity, oncogenic potential, immune-responsiveness, and germline mutation. In this review, we discuss advances in the field and the push toward clinical translation of IUGT.
Collapse
Affiliation(s)
- Citra N. Z. Mattar
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- National University Health SystemsSingaporeSingapore
| | - Jerry K. Y. Chan
- KK Women's and Children's HospitalSingaporeSingapore
- Duke‐NUS Medical SchoolSingaporeSingapore
| | - Mahesh Choolani
- Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- National University Health SystemsSingaporeSingapore
| |
Collapse
|
32
|
Moskowitzova K, Whitlock AE, Zurakowski D, Fauza DO. Hematogenous Routing of Exogenous mRNA Delivered Into the Amniotic Fluid. J Surg Res 2023; 289:116-120. [PMID: 37104922 DOI: 10.1016/j.jss.2023.03.037] [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: 09/14/2022] [Revised: 03/21/2023] [Accepted: 03/25/2023] [Indexed: 04/29/2023]
Abstract
INTRODUCTION Therapies based on exogenous messenger RNA (mRNA) administration have emerged as a powerful novel strategy for the actual or potential treatment of an assortment of diseases, including congenital surgical pathologies. We sought to determine whether the minimally invasive transamniotic route could be an alternative for prenatal mRNA delivery. METHODS Pregnant Sprague-Dawley dams underwent laparotomy followed by volume-matched intra-amniotic injections in all their fetuses (n = 120) of either a suspension of a custom firefly luciferase mRNA encapsulated by a lipid- and synthetic cationic polymer-based composite, or of a suspension of the same encapsulation components without mRNA, on gestational day 17 (E17; term = E21-22). On E18, E19, E20, and E21, samples from 14 fetal anatomical sites and maternal serum were procured for the screening of mRNA incorporation by host cells by measurement of luciferase activity via microplate luminometry. Statistical analysis was by Mann-Whitney U-test, including Bonferroni-adjustment. RESULTS Overall survival was 87.5% (105/120). Controlled by the encapsulating composite without mRNA, luciferase activity was detected in the animals that received encapsulated mRNA in the following fetal annexes: amniotic fluid, amnion, chorion, umbilical cord, and placenta (P = 0.033 to <0.001), as well as in the following fetal sites: liver, stomach, intestines, and lungs (P = 0.043-0.002). CONCLUSIONS Packaged exogenous mRNA can be incorporated by the fetus at least at select anatomical sites after simple intra-amniotic administration in a rodent model. The pattern and chronology of mRNA incorporation are compatible with transplacental hematogenous routing, as well as with fetal swallowing/aspiration. Further study of transamniotic mRNA administration is warranted.
Collapse
Affiliation(s)
- Kamila Moskowitzova
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, Massachusetts
| | - Ashlyn E Whitlock
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, Massachusetts
| | - David Zurakowski
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, Massachusetts
| | - Dario O Fauza
- Department of Surgery, Boston Children's Hospital/ Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
33
|
Singh K. Prenatal Interventions for the Treatment of Congenital Disorders. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
34
|
Berkowitz CL, Luks VL, Puc M, Peranteau WH. Molecular and Cellular In Utero Therapy. Clin Perinatol 2022; 49:811-820. [PMID: 36328600 DOI: 10.1016/j.clp.2022.06.005] [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: 01/27/2023]
Abstract
Significant advances in maternal-fetal medicine and gene sequencing technology have fostered a new frontier of in utero molecular and cellular therapeutics, including gene editing, enzyme replacement therapy, and stem cell transplantation to treat single-gene disorders with limited postnatal treatment strategies. In utero therapies take advantage of unique developmental properties of the fetus to allow for the correction of monogenic disorders before irreversible disease pathology develops. While early preclinical studies in animal models are encouraging, more studies are needed to further evaluate their safety and efficacy prior to widespread clinical use.
Collapse
Affiliation(s)
- Cara L Berkowitz
- Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Valerie L Luks
- Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Marcelina Puc
- Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - William H Peranteau
- Division of Pediatric General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
35
|
Huang K, Zapata D, Tang Y, Teng Y, Li Y. In vivo delivery of CRISPR-Cas9 genome editing components for therapeutic applications. Biomaterials 2022; 291:121876. [PMID: 36334354 PMCID: PMC10018374 DOI: 10.1016/j.biomaterials.2022.121876] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/15/2022] [Accepted: 10/23/2022] [Indexed: 12/07/2022]
Abstract
Since its mechanism discovery in 2012 and the first application for mammalian genome editing in 2013, CRISPR-Cas9 has revolutionized the genome engineering field and created countless opportunities in both basic science and translational medicine. The first clinical trial of CRISPR therapeutics was initiated in 2016, which employed ex vivo CRISPR-Cas9 edited PD-1 knockout T cells for the treatment of non-small cell lung cancer. So far there have been dozens of clinical trials registered on ClinicalTrials.gov in regard to using the CRISPR-Cas9 genome editing as the main intervention for therapeutic applications; however, most of these studies use ex vivo genome editing approach, and only a few apply the in vivo editing strategy. Compared to ex vivo editing, in vivo genome editing bypasses tedious procedures related to cell isolation, maintenance, selection, and transplantation. It is also applicable to a wide range of diseases and disorders. The main obstacles to the successful translation of in vivo therapeutic genome editing include the lack of safe and efficient delivery system and safety concerns resulting from the off-target effects. In this review, we highlight the therapeutic applications of in vivo genome editing mediated by the CRISPR-Cas9 system. Following a brief introduction of the history, biology, and functionality of CRISPR-Cas9, we showcase a series of exemplary studies in regard to the design and implementation of in vivo genome editing systems that target the brain, inner ear, eye, heart, liver, lung, muscle, skin, immune system, and tumor. Current challenges and opportunities in the field of CRISPR-enabled therapeutic in vivo genome editing are also discussed.
Collapse
Affiliation(s)
- Kun Huang
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Daniel Zapata
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Yan Tang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Yamin Li
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
36
|
Dai L, Du L. Genes in pediatric pulmonary arterial hypertension and the most promising BMPR2 gene therapy. Front Genet 2022; 13:961848. [PMID: 36506323 PMCID: PMC9730536 DOI: 10.3389/fgene.2022.961848] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare but progressive and lethal vascular disease of diverse etiologies, mainly caused by proliferation of endothelial cells, smooth muscle cells in the pulmonary artery, and fibroblasts, which ultimately leads to right-heart hypertrophy and cardiac failure. Recent genetic studies of childhood-onset PAH report that there is a greater genetic burden in children than in adults. Since the first-identified pathogenic gene of PAH, BMPR2, which encodes bone morphogenetic protein receptor 2, a receptor in the transforming growth factor-β superfamily, was discovered, novel causal genes have been identified and substantially sharpened our insights into the molecular genetics of childhood-onset PAH. Currently, some newly identified deleterious genetic variants in additional genes implicated in childhood-onset PAH, such as potassium channels (KCNK3) and transcription factors (TBX4 and SOX17), have been reported and have greatly updated our understanding of the disease mechanism. In this review, we summarized and discussed the advances of genetic variants underlying childhood-onset PAH susceptibility and potential mechanism, and the most promising BMPR2 gene therapy and gene delivery approaches to treat childhood-onset PAH in the future.
Collapse
|
37
|
Mondal G, VanLith CJ, Nicolas CT, Thompson WS, Cao WS, Hillin L, Haugo BJ, Brien DRO, Kocher JP, Kaiser RA, Lillegard JB. Activation of homology-directed DNA repair plays key role in CRISPR-mediated genome correction. Gene Ther 2022; 30:386-397. [PMID: 36258038 DOI: 10.1038/s41434-022-00369-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/09/2022]
Abstract
Gene editing for the cure of inborn errors of metabolism (IEMs) has been limited by inefficiency of adult hepatocyte targeting. Here, we demonstrate that in utero CRISPR/Cas9-mediated gene editing in a mouse model of hereditary tyrosinemia type 1 provides stable cure of the disease. Following this, we performed an extensive gene expression analysis to explore the inherent characteristics of fetal/neonatal hepatocytes that make them more susceptible to efficient gene editing than adult hepatocytes. We showed that fetal and neonatal livers are comprised of proliferative hepatocytes with abundant expression of genes involved in homology-directed repair (HDR) of DNA double-strand breaks (DSBs), key for efficient gene editing by CRISPR/Cas9. We demonstrated the same is true of hepatocytes after undergoing a regenerative stimulus (partial hepatectomy), where post-hepatectomy cells show a higher efficiency of HDR and correction. Specifically, we demonstrated that HDR-related genome correction is most effective in the replicative phase, or S-phase, of an actively proliferating cell. In conclusion, this study shows that taking advantage of or triggering cell proliferation, specifically DNA replication in S-phase, may serve as an important tool to improve efficiency of CRISPR/Cas9-mediated genome editing in the liver and provide a curative therapy for IEMs in both children and adults.
Collapse
Affiliation(s)
| | | | - Clara T Nicolas
- Department of Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Surgery, University of Alabama Birmingham, Birmingham, AL, USA
| | - Whitney S Thompson
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - William S Cao
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Lori Hillin
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Daniel R O' Brien
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Jean-Pierre Kocher
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Robert A Kaiser
- Department of Surgery, Mayo Clinic, Rochester, MN, USA.,Midwest Fetal Care Center, Children's Hospital of Minnesota, Minneapolis, MN, USA
| | - Joseph B Lillegard
- Department of Surgery, Mayo Clinic, Rochester, MN, USA. .,Midwest Fetal Care Center, Children's Hospital of Minnesota, Minneapolis, MN, USA. .,Pediatric Surgical Associates, Minneapolis, MN, USA.
| |
Collapse
|
38
|
Herzeg A, Almeida-Porada G, Charo RA, David AL, Gonzalez-Velez J, Gupta N, Lapteva L, Lianoglou B, Peranteau W, Porada C, Sanders SJ, Sparks TN, Stitelman DH, Struble E, Sumner CJ, MacKenzie TC. Prenatal Somatic Cell Gene Therapies: Charting a Path Toward Clinical Applications (Proceedings of the CERSI-FDA Meeting). J Clin Pharmacol 2022; 62 Suppl 1:S36-S52. [PMID: 36106778 PMCID: PMC9547535 DOI: 10.1002/jcph.2127] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/24/2022] [Indexed: 01/19/2023]
Abstract
We are living in a golden age of medicine in which the availability of prenatal diagnosis, fetal therapy, and gene therapy/editing make it theoretically possible to repair almost any defect in the genetic code. Furthermore, the ability to diagnose genetic disorders before birth and the presence of established surgical techniques enable these therapies to be delivered safely to the fetus. Prenatal therapies are generally used in the second or early third trimester for severe, life-threatening disorders for which there is a clear rationale for intervening before birth. While there has been promising work for prenatal gene therapy in preclinical models, the path to a clinical prenatal gene therapy approach is complex. We recently held a conference with the University of California, San Francisco-Stanford Center of Excellence in Regulatory Science and Innovation, researchers, patient advocates, regulatory (members of the Food and Drug Administration), and other stakeholders to review the scientific background and rationale for prenatal somatic cell gene therapy for severe monogenic diseases and initiate a dialogue toward a safe regulatory path for phase 1 clinical trials. This review represents a summary of the considerations and discussions from these conversations.
Collapse
Affiliation(s)
- Akos Herzeg
- UCSF Center for Maternal-Fetal PrecisionMedicine, San Francisco, California, USA
- Department of Surgery, University of California, San Francisco, California, USA
- Department of Obstetrics and Gynecology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Graca Almeida-Porada
- Fetal Research and Therapy Program, Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, North Carolina, USA
- Wake Forest University, School of Medicine, Winston-Salem, North Carolina, USA
| | - R. Alta Charo
- University of Wisconsin Law School, Madison, Wisconsin, USA
| | - Anna L. David
- Elizabeth Garrett Anderson Institute for Women’s Health, University College London Medical School, London, UK
- National Institute for Health Research University College London Hospitals Biomedical Research Centre, London, UK
| | - Juan Gonzalez-Velez
- UCSF Center for Maternal-Fetal PrecisionMedicine, San Francisco, California, USA
- Department of Obstetrics and Gynecology, University of California, San Francisco, California, USA
| | - Nalin Gupta
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
- Brain Tumor Center, University of California San Francisco, San Francisco, California, USA
- Department of Pediatrics and Benioff Children’s Hospital, University of California San Francisco, San Francisco, California, USA
| | - Larissa Lapteva
- Office of Tissues and Advanced Therapies/Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Washington, DC, USA
| | - Billie Lianoglou
- UCSF Center for Maternal-Fetal PrecisionMedicine, San Francisco, California, USA
- Department of Surgery, University of California, San Francisco, California, USA
| | - William Peranteau
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Christopher Porada
- Fetal Research and Therapy Program, Wake Forest Institute for Regenerative Medicine, Wake Forest University, Winston Salem, North Carolina, USA
- Wake Forest University, School of Medicine, Winston-Salem, North Carolina, USA
| | - Stephan J. Sanders
- UCSF Center for Maternal-Fetal PrecisionMedicine, San Francisco, California, USA
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, California, USA
- Institute for Human Genetics, University of California, San Francisco, California, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, California, USA
| | - Teresa N. Sparks
- UCSF Center for Maternal-Fetal PrecisionMedicine, San Francisco, California, USA
- Department of Obstetrics and Gynecology, University of California, San Francisco, California, USA
| | - David H. Stitelman
- Yale University School of Medicine, Department of Surgery, Division of Pediatric Surgery, New Haven, CT, USA
| | - Evi Struble
- Office of Tissues and Advanced Therapies/Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Washington, DC, USA
| | - Charlotte J. Sumner
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tippi C. MacKenzie
- UCSF Center for Maternal-Fetal PrecisionMedicine, San Francisco, California, USA
- Department of Surgery, University of California, San Francisco, California, USA
- Department of Obstetrics and Gynecology, University of California, San Francisco, California, USA
- Department of Pediatrics and Benioff Children’s Hospital, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
39
|
Alapati D, Shaffer TH. Administration of Drugs/Gene Products to the Respiratory System: A Historical Perspective of the Use of Inert Liquids. Front Physiol 2022; 13:871893. [PMID: 35620598 PMCID: PMC9127416 DOI: 10.3389/fphys.2022.871893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/31/2022] [Indexed: 11/20/2022] Open
Abstract
The present review is a historical perspective of methodology and applications using inert liquids for respiratory support and as a vehicle to deliver biological agents to the respiratory system. As such, the background of using oxygenated inert liquids (considered a drug when used in the lungs) opposed to an oxygen-nitrogen gas mixture for respiratory support is presented. The properties of these inert liquids and the mechanisms of gas exchange and lung function alterations using this technology are described. In addition, published preclinical and clinical trial results are discussed with respect to treatment modalities for respiratory diseases. Finally, this forward-looking review provides a comprehensive overview of potential methods for administration of drugs/gene products to the respiratory system and potential biomedical applications.
Collapse
Affiliation(s)
- Deepthi Alapati
- Nemours Children’s Health, Wilmington, DE, United States
- Sidney Kimmel School of Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Thomas H. Shaffer
- Nemours Children’s Health, Wilmington, DE, United States
- Sidney Kimmel School of Medicine, Thomas Jefferson University, Philadelphia, PA, United States
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
40
|
Lederer CW, Koniali L, Buerki-Thurnherr T, Papasavva PL, La Grutta S, Licari A, Staud F, Bonifazi D, Kleanthous M. Catching Them Early: Framework Parameters and Progress for Prenatal and Childhood Application of Advanced Therapies. Pharmaceutics 2022; 14:pharmaceutics14040793. [PMID: 35456627 PMCID: PMC9031205 DOI: 10.3390/pharmaceutics14040793] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 01/19/2023] Open
Abstract
Advanced therapy medicinal products (ATMPs) are medicines for human use based on genes, cells or tissue engineering. After clear successes in adults, the nascent technology now sees increasing pediatric application. For many still untreatable disorders with pre- or perinatal onset, timely intervention is simply indispensable; thus, prenatal and pediatric applications of ATMPs hold great promise for curative treatments. Moreover, for most inherited disorders, early ATMP application may substantially improve efficiency, economy and accessibility compared with application in adults. Vindicating this notion, initial data for cell-based ATMPs show better cell yields, success rates and corrections of disease parameters for younger patients, in addition to reduced overall cell and vector requirements, illustrating that early application may resolve key obstacles to the widespread application of ATMPs for inherited disorders. Here, we provide a selective review of the latest ATMP developments for prenatal, perinatal and pediatric use, with special emphasis on its comparison with ATMPs for adults. Taken together, we provide a perspective on the enormous potential and key framework parameters of clinical prenatal and pediatric ATMP application.
Collapse
Affiliation(s)
- Carsten W. Lederer
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.K.); (P.L.P.); (M.K.)
- Correspondence: ; Tel.: +357-22-392764
| | - Lola Koniali
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.K.); (P.L.P.); (M.K.)
| | - Tina Buerki-Thurnherr
- Empa, Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland;
| | - Panayiota L. Papasavva
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.K.); (P.L.P.); (M.K.)
| | - Stefania La Grutta
- Institute of Translational Pharmacology, IFT National Research Council, 90146 Palermo, Italy;
| | - Amelia Licari
- Pediatric Clinic, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy;
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, 50005 Hradec Králové, Czech Republic;
| | - Donato Bonifazi
- Consorzio per Valutazioni Biologiche e Farmacologiche (CVBF) and European Paediatric Translational Research Infrastructure (EPTRI), 70122 Bari, Italy;
| | - Marina Kleanthous
- The Molecular Genetics Thalassemia Department, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus; (L.K.); (P.L.P.); (M.K.)
| |
Collapse
|
41
|
Xu Z, Wang Q, Zhong H, Jiang Y, Shi X, Yuan B, Yu N, Zhang S, Yuan X, Guo S, Yang Y. Carrier strategies boost the application of CRISPR/Cas system in gene therapy. EXPLORATION (BEIJING, CHINA) 2022; 2:20210081. [PMID: 37323878 PMCID: PMC10190933 DOI: 10.1002/exp.20210081] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/06/2022] [Indexed: 02/05/2023]
Abstract
Emerging clustered regularly interspaced short palindromic repeat/associated protein (CRISPR/Cas) genome editing technology shows great potential in gene therapy. However, proteins and nucleic acids suffer from enzymatic degradation in the physiological environment and low permeability into cells. Exploiting carriers to protect the CRISPR system from degradation, enhance its targeting of specific tissues and cells, and reduce its immunogenicity is essential to stimulate its clinical applications. Here, the authors review the state-of-the-art CRISPR delivery systems and their applications, and describe strategies to improve the safety and efficacy of CRISPR mediated genome editing, categorized by three types of cargo formats, that is, Cas: single-guide RNA ribonucleoprotein, Cas mRNA and single-guide RNA, and Cas plasmid expressing CRISPR/Cas systems. The authors hope this review will help develop safe and efficient nanomaterial-based carriers for CRISPR tools.
Collapse
Affiliation(s)
- Zunkai Xu
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Qingnan Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation CenterChengduChina
| | - Haiping Zhong
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Yaoyao Jiang
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Xiaoguang Shi
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Bo Yuan
- School of MedicineNankai UniversityTianjinChina
- Tianjin Key Laboratory of Ophthalmology and Visual ScienceTianjin Eye InstituteTianjin Eye HospitalTianjinChina
| | - Na Yu
- Translational Medicine CenterKey Laboratory of Molecular Target & Clinical PharmacologySchool of Pharmaceutical Sciences and The Second Affiliated HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of EducationDalian Minzu UniversityDalianChina
| | - Xiaoyong Yuan
- Tianjin Key Laboratory of Ophthalmology and Visual ScienceTianjin Eye InstituteTianjin Eye HospitalTianjinChina
- Clinical College of OphthalmologyTianjin Medical UniversityTianjinChina
| | - Shutao Guo
- Key Laboratory of Functional Polymer Materials of Ministry of EducationState Key Laboratory of Medicinal Chemical Biology and Institute of Polymer ChemistryCollege of ChemistryNankai UniversityTianjinChina
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation CenterChengduChina
| |
Collapse
|
42
|
Bisserier M, Sun XQ, Fazal S, Turnbull IC, Bonnet S, Hadri L. Novel Insights into the Therapeutic Potential of Lung-Targeted Gene Transfer in the Most Common Respiratory Diseases. Cells 2022; 11:984. [PMID: 35326434 PMCID: PMC8947048 DOI: 10.3390/cells11060984] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 12/10/2022] Open
Abstract
Over the past decades, a better understanding of the genetic and molecular alterations underlying several respiratory diseases has encouraged the development of new therapeutic strategies. Gene therapy offers new therapeutic alternatives for inherited and acquired diseases by delivering exogenous genetic materials into cells or tissues to restore physiological protein expression and/or activity. In this review, we review (1) different types of viral and non-viral vectors as well as gene-editing techniques; and (2) the application of gene therapy for the treatment of respiratory diseases and disorders, including pulmonary arterial hypertension, idiopathic pulmonary fibrosis, cystic fibrosis, asthma, alpha-1 antitrypsin deficiency, chronic obstructive pulmonary disease, non-small-cell lung cancer, and COVID-19. Further, we also provide specific examples of lung-targeted therapies and discuss the major limitations of gene therapy.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| | - Xiao-Qing Sun
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Shahood Fazal
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| | - Irene C. Turnbull
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| | - Sébastien Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec, QC G1V4G5, Canada;
- Department of Medicine, Laval University, Québec, QC G1V4G5, Canada
| | - Lahouaria Hadri
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (M.B.); (S.F.); (I.C.T.)
| |
Collapse
|
43
|
Rasul MF, Hussen BM, Salihi A, Ismael BS, Jalal PJ, Zanichelli A, Jamali E, Baniahmad A, Ghafouri-Fard S, Basiri A, Taheri M. Strategies to overcome the main challenges of the use of CRISPR/Cas9 as a replacement for cancer therapy. Mol Cancer 2022; 21:64. [PMID: 35241090 PMCID: PMC8892709 DOI: 10.1186/s12943-021-01487-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/26/2021] [Indexed: 12/11/2022] Open
Abstract
CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats-associated protein 9) shows the opportunity to treat a diverse array of untreated various genetic and complicated disorders. Therapeutic genome editing processes that target disease-causing genes or mutant genes have been greatly accelerated in recent years as a consequence of improvements in sequence-specific nuclease technology. However, the therapeutic promise of genome editing has yet to be explored entirely, many challenges persist that increase the risk of further mutations. Here, we highlighted the main challenges facing CRISPR/Cas9-based treatments and proposed strategies to overcome these limitations, for further enhancing this revolutionary novel therapeutics to improve long-term treatment outcome human health.
Collapse
Affiliation(s)
- Mohammed Fatih Rasul
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Kurdistan region, Erbil, Iraq.,Center of Research and Strategic Studies, Lebanese French University, Erbil, Iraq
| | - Abbas Salihi
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Iraq.,Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Iraq
| | - Bnar Saleh Ismael
- Department of Pharmacology and Toxicology, College of Pharmacy, Hawler Medical University, Kurdistan region, Erbil, Iraq
| | - Paywast Jamal Jalal
- Biology Department, College of Science, University of Sulaimani, Sulaimani, Iraq
| | - Anna Zanichelli
- Department of Biomedical Sciences, University of Westminster, London, UK
| | - Elena Jamali
- Department of Pathology, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany. .,Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
White BM, Morrisey EE, Peranteau WH. In Utero Gene Editing for Inherited Lung Diseases. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-021-00205-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
45
|
Cooney AL, Wambach JA, Sinn PL, McCray PB. Gene Therapy Potential for Genetic Disorders of Surfactant Dysfunction. Front Genome Ed 2022; 3:785829. [PMID: 35098209 PMCID: PMC8798122 DOI: 10.3389/fgeed.2021.785829] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/15/2021] [Indexed: 12/30/2022] Open
Abstract
Pulmonary surfactant is critically important to prevent atelectasis by lowering the surface tension of the alveolar lining liquid. While respiratory distress syndrome (RDS) is common in premature infants, severe RDS in term and late preterm infants suggests an underlying genetic etiology. Pathogenic variants in the genes encoding key components of pulmonary surfactant including surfactant protein B (SP-B, SFTPB gene), surfactant protein C (SP-C, SFTPC gene), and the ATP-Binding Cassette transporter A3 (ABCA3, ABCA3 gene) result in severe neonatal RDS or childhood interstitial lung disease (chILD). These proteins play essential roles in pulmonary surfactant biogenesis and are expressed in alveolar epithelial type II cells (AEC2), the progenitor cell of the alveolar epithelium. SP-B deficiency most commonly presents in the neonatal period with severe RDS and requires lung transplantation for survival. SFTPC mutations act in an autosomal dominant fashion and more commonly presents with chILD or idiopathic pulmonary fibrosis than neonatal RDS. ABCA3 deficiency often presents as neonatal RDS or chILD. Gene therapy is a promising option to treat monogenic lung diseases. Successes and challenges in developing gene therapies for genetic disorders of surfactant dysfunction include viral vector design and tropism for target cell types. In this review, we explore adeno-associated virus (AAV), lentiviral, and adenoviral (Ad)-based vectors as delivery vehicles. Both gene addition and gene editing strategies are compared to best design treatments for lung diseases resulting from pathogenic variants in the SFTPB, SFTPC, and ABCA3 genes.
Collapse
Affiliation(s)
- Ashley L. Cooney
- Department of Pediatrics, The University of Iowa, Iowa City, IA, United States
- Pappajohn Biomedical Institute and the Center for Gene Therapy, The University of Iowa, Iowa City, IA, United States
- *Correspondence: Ashley L. Cooney,
| | - Jennifer A. Wambach
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Patrick L. Sinn
- Department of Pediatrics, The University of Iowa, Iowa City, IA, United States
- Pappajohn Biomedical Institute and the Center for Gene Therapy, The University of Iowa, Iowa City, IA, United States
| | - Paul B. McCray
- Department of Pediatrics, The University of Iowa, Iowa City, IA, United States
- Pappajohn Biomedical Institute and the Center for Gene Therapy, The University of Iowa, Iowa City, IA, United States
| |
Collapse
|
46
|
Swingle KL, Billingsley MM, Bose SK, White B, Palanki R, Dave A, Patel SK, Gong N, Hamilton AG, Alameh MG, Weissman D, Peranteau WH, Mitchell MJ. Amniotic fluid stabilized lipid nanoparticles for in utero intra-amniotic mRNA delivery. J Control Release 2022; 341:616-633. [PMID: 34742747 PMCID: PMC8776620 DOI: 10.1016/j.jconrel.2021.10.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/03/2021] [Accepted: 10/31/2021] [Indexed: 01/03/2023]
Abstract
Congenital disorders resulting in pathological protein deficiencies are most often treated postnatally with protein or enzyme replacement therapies. However, treatment of these disorders in utero before irreversible disease onset could significantly minimize disease burden, morbidity, and mortality. One possible strategy for the prenatal treatment of congenital disorders is in utero delivery of messenger RNA (mRNA). mRNA is a nucleic acid therapeutic that has previously been investigated as a platform for protein replacement therapies and gene editing technologies. While viral vectors have been explored to induce intracellular expression of mRNA, they are limited in their clinical application due to risks associated with immunogenicity and genomic integration. As an alternative to viral vectors, safe and efficient in utero mRNA delivery can be achieved using ionizable lipid nanoparticles (LNPs). While LNPs have demonstrated potent in vivo mRNA delivery to the liver following intravenous administration, intra-amniotic delivery has the potential to deliver mRNA to cells and tissues beyond those in the liver, such as in the skin, lung, and digestive tract. However, LNP stability in fetal amniotic fluid and how this stability affects mRNA delivery has not been previously investigated. Here, we engineered a library of LNPs using orthogonal design of experiments (DOE) to evaluate how LNP structure affects their stability in amniotic fluid ex utero and whether a lead candidate identified from these stability measurements enables intra-amniotic mRNA delivery in utero. We used a combination of techniques including dynamic light scattering (DLS), transmission electron microscopy (TEM), and chromatography followed by protein content quantification to screen LNP stability in amniotic fluids. These results identified multiple lead LNP formulations that are highly stable in amniotic fluids ranging from small animals to humans, including mouse, sheep, pig, and human amniotic fluid samples. We then demonstrate that stable LNPs from the ex utero screen in mouse amniotic fluid enabled potent mRNA delivery in primary fetal lung fibroblasts and in utero following intra-amniotic injection in a murine model. This exploration of ex utero stability in amniotic fluids demonstrates a means by which to identify novel LNP formulations for prenatal treatment of congenital disorders via in utero mRNA delivery.
Collapse
Affiliation(s)
- Kelsey L. Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Sourav K. Bose
- The Center for Fetal Research, Division of Pediatric General, Thoracic, and Fetal Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brandon White
- The Center for Fetal Research, Division of Pediatric General, Thoracic, and Fetal Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Rohan Palanki
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.,The Center for Fetal Research, Division of Pediatric General, Thoracic, and Fetal Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Apeksha Dave
- The Center for Fetal Research, Division of Pediatric General, Thoracic, and Fetal Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Savan K. Patel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alex G. Hamilton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Drew Weissman
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William H. Peranteau
- The Center for Fetal Research, Division of Pediatric General, Thoracic, and Fetal Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.,Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
47
|
Shanahan MA, Aagaard KM, McCullough LB, Chervenak FA, Shamshirsaz AA. Society for Maternal-Fetal Medicine Special Statement: Beyond the scalpel: in utero fetal gene therapy and curative medicine. Am J Obstet Gynecol 2021; 225:B9-B18. [PMID: 34537158 DOI: 10.1016/j.ajog.2021.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
With the recent advances in gene editing with systems such as CRISPR-Cas9, precise genome editing in utero is on the horizon. Sickle cell disease is an excellent candidate for in utero fetal gene therapy, because the disease is monogenic, causes irreversible harm, and has life-limiting morbidity. Gene therapy has recently been proven to be effective in an adolescent patient. Several hurdles still impede the progress for fetal gene therapy in humans, including an incomplete understanding of the fetal immune system, unclear maternal immune responses to in utero gene therapy, risks of off-target effects from gene editing, gestational age constraints, and ethical questions surrounding fetal genetic intervention. However, none of these barriers appears insurmountable, and the journey to in utero gene therapy for sickle cell disease and other conditions should be well underway.
Collapse
Affiliation(s)
- Matthew A Shanahan
- Society for Maternal-Fetal Medicine, 409 12 St. SW, Washington, DC 20024, USA.
| | - Kjersti M Aagaard
- Society for Maternal-Fetal Medicine, 409 12 St. SW, Washington, DC 20024, USA.
| | | | - Francis A Chervenak
- Society for Maternal-Fetal Medicine, 409 12 St. SW, Washington, DC 20024, USA.
| | | |
Collapse
|
48
|
Abstract
Prenatal gene therapy could provide a cure for many monogenic diseases. Prenatal gene therapy has multiple potential advantages over postnatal therapy, including treating before the onset of disease, the ability to induce tolerance and cross the blood-brain barrier. In this chapter, we will describe in utero gene therapy and its rationale, clinical trials of postnatal gene therapy, preclinical studies of in utero gene therapy, and potential risks to the mother and fetus.
Collapse
Affiliation(s)
- Marisa E Schwab
- Center for Maternal-Fetal Precision Medicine
- Department of Surgery, University of California, San Francisco, San Francisco, California
| | - Tippi C MacKenzie
- Center for Maternal-Fetal Precision Medicine
- Department of Surgery, University of California, San Francisco, San Francisco, California
| |
Collapse
|
49
|
Abstract
Gene therapy has traditionally involved the delivery of exogenous genetic material to a cell-most commonly to replace defective genes causing monogenic disorders. This allows cells to produce proteins that are otherwise absent in sufficient quantities, ideally for a therapeutic purpose. Since its inception over 40 years ago, the field of gene therapy has significantly expanded and now includes targeted gene editing strategies, including, but not limited to, clustered regularly interspaced palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9), transcription activator-like effector nucleases (TALENs), and zinc-finger nucleases (ZFNs).
Collapse
Affiliation(s)
- Barbara Coons
- Center for Fetal Research, Division of General, Thoracic, and Fetal Surgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | |
Collapse
|
50
|
Luthra R, Kaur S, Bhandari K. Applications of CRISPR as a potential therapeutic. Life Sci 2021; 284:119908. [PMID: 34453943 DOI: 10.1016/j.lfs.2021.119908] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 02/06/2023]
Abstract
Genetic disorders and congenital abnormalities are present in 2-5% of births all over the world and can cause up to 50% of all early childhood deaths. The establishment of sophisticated and controlled techniques for customizing DNA manipulation is significant for the therapeutic role in such disorders and further research on them. One such technique is CRISPR that is significant towards optimizing genome editing and therapies, metabolic fluxes as well as artificial genetic systems. CRISPR-Cas9 is a molecular appliance that is applied in the areas of genetic and protein engineering. The CRISPR-CAS system is an integral element of prokaryotic adaptive immunity that allows prokaryotic cells to identify and kill any foreign DNA. The Gene editing property of CRISPR finds various applications like diagnostics and therapeutics in cancer, neurodegenerative disorders, genetic diseases, blindness, etc. This review discusses applications of CRISPR as a therapeutic in various disorders including several genetic diseases (including sickle cell anemia, blindness, thalassemia, cystic fibrosis, hereditary tyrosinemia type I, duchenne muscular dystrophy, mitochondrial disorders), Cancer, Huntington's disease and viral infections (like HIV, COVID, etc.) along with the prospects concerning them. CRISPR-based therapy is also being researched and defined for COVID-19. The related mechanism of CRISPR has been discussed alongside highlighting challenges involved in therapeutic applications of CRISPR.
Collapse
Affiliation(s)
- Ritika Luthra
- Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Simran Kaur
- Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Kriti Bhandari
- Department of Biotechnology, Delhi Technological University, Delhi, India.
| |
Collapse
|