51
|
Xiong Q, Zhu C, Yin X, Zhu L. CRISPR/Cas and Argonaute-based biosensors for nucleic acid detection. Talanta 2025; 294:128210. [PMID: 40280080 DOI: 10.1016/j.talanta.2025.128210] [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: 03/06/2025] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Nowadays, nucleic acid detection technology has been applied to disease diagnosis, prevention, food safety, environmental testing and many other aspects. However, traditional methods still have shortcomings. Therefore, there is an urgent need for a simple, rapid, sensitive, and specific new method to supersede traditional nucleic acid detection technology. CRISPR/Cas(Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated) system and Argonaute (Ago) system play an important role in microbial immune defense. Their targeting specificity, programmability and special trans-cleavage activity make it possible to develop some new platforms for nucleic acid detection in combination with a variety of biosensors. We introduce the origins of these two systems and the biosensors developed based on CRISPR/Cas system and Ago system, respectively, especially the prospects for the future development of Cascade Amplification biosensors. This review is expected to provide useful guidance for researchers in related fields and provide inspiration for the development of Cascade Amplification biosensors in the future.
Collapse
Affiliation(s)
- Qiangyuan Xiong
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China; University of Science and Technology of China, Hefei, 230026, PR China
| | - Cancan Zhu
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China.
| | - Xueer Yin
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China; University of Science and Technology of China, Hefei, 230026, PR China
| | - Ling Zhu
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China; University of Science and Technology of China, Hefei, 230026, PR China.
| |
Collapse
|
52
|
Zhao C, Cao Y, Ibrahim N, Wang Y, Martemyanov KA. Efficient in vivo labeling of endogenous proteins with SMART delineates retina cellular and synaptic organization. Nat Commun 2025; 16:3768. [PMID: 40263339 PMCID: PMC12015494 DOI: 10.1038/s41467-025-58945-6] [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: 12/27/2023] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
A key application of CRISPR/Cas9-based genomic editing is modification of genes to introduce engineered sequences. However, the editing flexibility is severely constrained by the requirement for targeting sites in proximity to the desired modification site, which makes many modifications intractable. Here, we develop a strategy that overcomes this key limitation to allow CRISPR-based editing at any position with high efficiency. It relies on reconstructing the targeted gene using Silently Mutate And Repair Template (SMART) where we mutate the gap sequence in the repair template to prevent its base pairing with the target DNA while maintaining the same amino acid coding. Using vertebrate retina as a neuronal model system we document the application of SMART editing for labeling endogenous proteins in vivo with high efficiency. We show that SMART editing allows us to access numerous cell types in the retina and address fundamental cell biological questions pertaining to its organization. We propose that this approach will facilitate functional genomic studies in a wide range of systems and increase the precision of corrective gene therapies.
Collapse
Affiliation(s)
- Chuanping Zhao
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
- Skaggs Graduate School, The Scripps Research Institute, Jupiter, FL, USA
| | - Yan Cao
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Noor Ibrahim
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
| | - Yuchen Wang
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA.
- Skaggs Graduate School, The Scripps Research Institute, Jupiter, FL, USA.
| |
Collapse
|
53
|
Thornburg CD, Pipe SW, Cantore A, Unzu C, Jones M, Miesbach WA. Clinical perspective: Advancing hemophilia treatment through gene therapy approaches. Mol Ther 2025:S1525-0016(25)00297-7. [PMID: 40263938 DOI: 10.1016/j.ymthe.2025.04.023] [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: 12/23/2024] [Revised: 03/21/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025] Open
Abstract
Hemophilia, a congenital bleeding disorder, can cause arthropathy, impaired mobility, pain, and life-threatening hemorrhage events, significantly impacting quality of life for patients and caregivers. Current therapies, although effective, necessitate costly lifelong treatment, often in specialized settings. However, as a monogenic disorder caused by loss-of-function genetic variants, hemophilia is amenable to gene therapy. In this article, three primary gene therapy approaches at the forefront of clinical development are reviewed. Adeno-associated virus-based gene therapy, having secured approval in the EU, UK, and US after promising phase 3 trial results, demonstrates clear superiority over standard-of-care treatment. Lentivirus-based approaches capable of transducing dividing and nondividing cells may improve the durability of treatment and have low susceptibility to pre-existing neutralizing antibodies to viral vectors. Finally, gene editing techniques such as zinc finger nucleases and CRISPR aim to correct genetic defects directly, holding promise as novel, effective, and highly durable therapeutic strategies in adults and children with hemophilia. This review provides a comprehensive summary of the current status of these gene therapy approaches, highlighting advantages, limitations, and potential future developments.
Collapse
Affiliation(s)
- Courtney D Thornburg
- National Institutes of Health, National Heart, Lung, and Blood Institute, Division of Blood Diseases and Resources, Bethesda, MD, USA
| | - Steve W Pipe
- Pediatric Hematology-Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Alessio Cantore
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Carmen Unzu
- DNA and RNA Medicine Division, CIMA Universidad de Navarra, Pamplona, Spain
| | | | - Wolfgang A Miesbach
- Department of Haemostaseology University Hospital Frankfurt, Frankfurt, Germany.
| |
Collapse
|
54
|
Said N, Venketaraman V. Neuroinflammation, Blood-Brain Barrier, and HIV Reservoirs in the CNS: An In-Depth Exploration of Latency Mechanisms and Emerging Therapeutic Strategies. Viruses 2025; 17:572. [PMID: 40285014 PMCID: PMC12030944 DOI: 10.3390/v17040572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/12/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025] Open
Abstract
Despite the success of antiretroviral therapy (ART) in suppressing viral replication in the blood, HIV persists in the central nervous system (CNS) and causes chronic neurocognitive impairment, a hallmark of HIV-associated neurocognitive disorders (HAND). This review looks at the complex interactions among HIV, the blood-brain barrier (BBB), neuroinflammation, and the roles of viral proteins, immune cell trafficking, and pro-inflammatory mediators in establishing and maintaining latent viral reservoirs in the CNS, particularly microglia and astrocytes. Key findings show disruption of the BBB, monocyte infiltration, and activation of CNS-resident cells by HIV proteins like Tat and gp120, contributing to the neuroinflammatory environment and neuronal damage. Advances in epigenetic regulation of latency have identified targets like histone modifications and DNA methylation, and new therapeutic strategies like latency-reversing agents (LRAs), gene editing (CRISPR/Cas9), and nanoparticle-based drug delivery also offer hope. While we have made significant progress in understanding the molecular basis of HIV persistence in the CNS, overcoming the challenges of BBB penetration and neuroinflammation is key to developing effective therapies. Further research into combination therapies and novel drug delivery systems will help improve outcomes for HAND patients and bring us closer to a functional cure for HIV.
Collapse
Affiliation(s)
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA;
| |
Collapse
|
55
|
Lu P, Long J. SOHO State of the Art Updates and Next Questions | CD7 CAR-T Therapy for Treating CD7-Positive Hematological Malignancies: Clinical Advances and Future Directions. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025:S2152-2650(25)00142-9. [PMID: 40374439 DOI: 10.1016/j.clml.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 05/17/2025]
Abstract
CD7 CAR-T cell therapy has emerged as a promising treatment for relapsed/refractory (R/R) CD7-positive hematological malignancies, offering new hope for patients with limited therapeutic options. This review examines the recent clinical advances, challenges, and future directions of CD7 CAR-T therapy. Clinical trials have demonstrated remarkable efficacy of CD7 chimeric antigen receptor T (CD7 CAR-T) cells in treating T-cell acute lymphoblastic leukemia (T-ALL), T-cell lymphoblastic lymphoma (T-LBL), and other CD7-positive malignancies, with complete remission (CR) rates of 90-95% in bone marrow (BM) and 50% to 60% in extramedullary disease (EMD). Various engineering strategies, including naturally selected CD7-targeted CAR-T cells, gene editing, protein blockers and universal CAR-T cells, have been developed to overcome challenges such as fratricide. While CD7 CAR-T therapy has shown promising initial responses, durable remissions often depend on consolidative allogeneic hematopoietic stem cell transplantation (allo-HSCT). Ongoing research is focused on optimizing CAR designs, improving CAR-T cell persistence, and developing novel combination strategies to enhance long-term outcomes. Safety profiles have been generally manageable, with cytokine release syndrome (CRS) and neurotoxicity being the primary concerns. However, prolonged cytopenias and potential long-term immunodeficiency due to depletion of healthy CD7-positive cells remain areas of active investigation. As CD7 CAR-T therapy continues to evolve, future directions include refining patient selection, exploring dual-targeting approaches, and investigating innovative strategies to integrate CAR-T therapy with allo-HSCT. These advancements aim to improve the efficacy, safety, and accessibility of CD7 CAR-T therapy for patients with CD7-positive hematological malignancies.
Collapse
Affiliation(s)
- Peihua Lu
- Hebei Yanda Lu Daopei Hospital, Langfang, China; Beijing Lu Daopei Hospital, Beijing, China; Beijing Lu Daopei Institute of Hematology, Beijing, China.
| | - Jing Long
- Beijing Lu Daopei Institute of Hematology, Beijing, China
| |
Collapse
|
56
|
Shanley HT, Wang T, Taki AC, Byrne JJ, Chang BCH, Sleebs BE, Gasser RB. Advances in Anthelmintic Target Identification. Int J Mol Sci 2025; 26:3738. [PMID: 40332360 PMCID: PMC12028019 DOI: 10.3390/ijms26083738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 05/08/2025] Open
Abstract
Parasitic nematodes pose a significant threat to human and animal health, causing widespread morbidity and substantial socioeconomic losses globally. Despite the utility of anthelmintic drugs in parasite control, the emergence of widespread resistance necessitates the discovery of novel interventions. Advances through the use of whole-organism phenotypic screening have identified some promising nematocidal compounds, including nemacol, tolfenpyrad, UMW-9729, and ABX464. This article summarises efforts in this discovery, with a focus on Haemonchus contortus and Caenorhabditis elegans as model nematodes, and discusses approaches used for drug target deconvolution, including proteomic, chemical and genetic/genomic techniques. Stability-based proteomic assays, such as thermal proteome profiling, have been useful for identifying protein targets for these compounds, shedding light on their mechanisms of action. However, challenges remain in extrapolating findings from C. elegans to parasitic nematodes, emphasising the need for validation studies. Understanding drug-target interactions in nematodes is critical for developing next-generation anthelmintics and for mitigating the growing resistance challenge. This review outlines recent progress in this area and discusses future directions in target validation and anthelmintic development to support parasite control programmes.
Collapse
Affiliation(s)
- Harrison T. Shanley
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia; (H.T.S.); (T.W.); (A.C.T.); (J.J.B.); (B.C.H.C.)
| | - Tao Wang
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia; (H.T.S.); (T.W.); (A.C.T.); (J.J.B.); (B.C.H.C.)
| | - Aya C. Taki
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia; (H.T.S.); (T.W.); (A.C.T.); (J.J.B.); (B.C.H.C.)
| | - Joseph J. Byrne
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia; (H.T.S.); (T.W.); (A.C.T.); (J.J.B.); (B.C.H.C.)
| | - Bill C. H. Chang
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia; (H.T.S.); (T.W.); (A.C.T.); (J.J.B.); (B.C.H.C.)
| | - Brad E. Sleebs
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia; (H.T.S.); (T.W.); (A.C.T.); (J.J.B.); (B.C.H.C.)
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Robin B. Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC 3010, Australia; (H.T.S.); (T.W.); (A.C.T.); (J.J.B.); (B.C.H.C.)
| |
Collapse
|
57
|
Towell SE, Jareczek MJ, Cooke LS, Godfrey DR, Zhukhovitskiy AV. Skeletal Editing of Polymer Backbones and Its Impact Across the Polymer Lifecycle. Acc Chem Res 2025; 58:1275-1283. [PMID: 40173419 DOI: 10.1021/acs.accounts.5c00054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
ConspectusIn the last five years, interest in the precise modification of molecular cores─termed skeletal editing─has rapidly expanded in the Chemistry community. Beyond the intrinsic value of these transformations, skeletal editing also has value in the attention it brings to under-explored chemical challenges, whose solutions could transform the practice of Chemistry at large. In few contexts does this perspective ring as true as in the realm of polymers. Inspired by the revolutionary power of biologically derived machinery called CRISPR-Cas9 to edit nucleic acid polymers and, consequently, the genetic meaning encoded in them, we envisioned that skeletal editing of synthetic polymer backbones may also enable control over the structure and "meaning"─i.e., properties and function─of plastics. However, the idea of editing polymer backbones brings about numerous fundamental chemical questions that must be answered to make the vision a reality: for instance, how to constructively activate carbon-carbon and carbon-heteroatom bonds that make up typical polymer backbones and how to do so in a site-selective manner? While many fundamental questions have begun to be answered by the small molecule community, they are yet to be applied to the realm of polymers, and such adaptation often begets new scientific challenges. Moreover, as we begin to tackle these questions, we must always consider how advances in skeletal editing of polymer backbones impact the broader contexts of applications and sustainability of plastics.In this Account, we summarize our efforts to advance the skeletal editing of polymer backbones, focusing on how such methods can affect each stage of the polymer lifecycle: (1) provide an entry to previously challenging-to-access functional polymers or to existing ones but from new feedstocks, (2) evolve one type of polymer into another with associated changes in material properties, and (3) enable the breakdown of otherwise intractable polymer backbones. Along the way, we describe our rationale behind the selection and development of reactions utilized for skeletal editing. We explain how small molecule reactions often need to be adapted to suit polymeric substrates and the methodology optimizations we needed to do to accomplish our edits. We also discuss the considerations involved in the selection or design of polymeric substrates for editing with an eye toward what edits can add to polymer function and how to advance the field. We conclude with an outlook on outstanding challenges that we aim to address in future work establishing areas for future exploration within each of our topic areas.
Collapse
Affiliation(s)
- Sydney E Towell
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mark J Jareczek
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Lauren S Cooke
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Daniel R Godfrey
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Aleksandr V Zhukhovitskiy
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
58
|
Chen WD, Liu L, Cheng L. Functionally Tunable Star-Shaped Multivalent crRNAs for Photocontrol CRISPR/Cas Editing. Angew Chem Int Ed Engl 2025:e202506527. [PMID: 40227971 DOI: 10.1002/anie.202506527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 04/16/2025]
Abstract
Clustered regularly interspaced shortpalindromic repeats/CRISPR-associated (CRISPR/Cas)-based genome editing has significantly advanced genetic engineering due to its precision, simplicity, and versatility. However, achieving precise spatial and temporal control remains challenging, restricting therapeutic and research applications. Herein, we introduce a novel class of star-shaped, multivalent crRNAs engineered for precise spatiotemporal control of CRISPR/Cas9 and Cas12a editing systems. These crRNAs are synthesized via single-site chemical modification and can be efficiently purified. By integrating distinct photo-responsive chemical linkages, we achieved selective activation of crRNA activity upon irradiation with specific wavelengths, enabling orthogonal regulation of multiple genetic targets simultaneously. This method demonstrated robust OFF-ON switching capabilities in vitro, characterized by minimal leakage and rapid activation. Importantly, the approach also proved highly effective for temporally controlled gene editing in mammalian cells in vivo, achieving considerable editing efficiency following brief photoactivation. Due to its target sequence-independent, single-site modification design, this strategy may serve as a universal solution for diverse CRISPR/Cas systems, eliminating cumbersome optimization processes. Future advancements incorporating long-wavelength responsive and reversible linkers promise further enhancement of tissue penetration and control, significantly broadening the applicability and impact of this approach in biological research and therapeutic interventions.
Collapse
Affiliation(s)
- Wen-Da Chen
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Laboratory of Molecular Recognition and Function, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Li Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Laboratory of Molecular Recognition and Function, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liang Cheng
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Laboratory of Molecular Recognition and Function, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
59
|
Zheng M, Bao N, Wang Z, Song C, Jin Y. Alternative splicing in autism spectrum disorder: Recent insights from mechanisms to therapy. Asian J Psychiatr 2025; 108:104501. [PMID: 40273800 DOI: 10.1016/j.ajp.2025.104501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025]
Abstract
Alternative splicing (AS) is a vital and highly dynamic RNA regulatory mechanism that allows a single gene to generate multiple mRNA and protein isoforms. Dysregulation of AS has been identified as a key contributor to the pathogenesis of autism spectrum disorders (ASD). A comprehensive understanding of aberrant splicing mechanisms and their functional consequences in ASD can help uncover the molecular basis of the disorder and facilitate the development of therapeutic strategies. This review focuses on the major aberrant splicing events and key splicing regulators associated with ASD, highlighting their roles in linking defective splicing to ASD pathogenesis. In addition, a discussion of how emerging technologies, such as long-read sequencing, single-cell sequencing, spatial transcriptomics and CRISPR-Cas systems are offering novel insights into the role and mechanisms of AS in ASD is presented. Finally, the RNA splicing-based therapeutic strategies are evaluated, emphasizing their potential to address unmet clinical needs in ASD treatment.
Collapse
Affiliation(s)
- Mixue Zheng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| | - Nengcheng Bao
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Zhechao Wang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, College of Life Sciences, Zhejiang University, Hangzhou 310058, China; School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Chao Song
- Department of Developmental and Behavioral Pediatrics, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Centre for Child Health, Hangzhou 310052, China.
| | - Yongfeng Jin
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
60
|
Santiago-Rivera E, Scheibel T. Spider Eye Development Editing and Silk Fiber Engineering Using CRISPR-Cas. Angew Chem Int Ed Engl 2025:e202502068. [PMID: 40223236 DOI: 10.1002/anie.202502068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/30/2025] [Accepted: 04/04/2025] [Indexed: 04/15/2025]
Abstract
CRISPR-Cas9 gene editing represents an effective and precise technology to induce mutations in the genome, and it has been applied to a wide range of organisms for diverse purposes. However, CRISPR-based gene editing in spiders has not been reported to date. In this study, we demonstrate CRISPR-mediated microinjection in parental spiders leading to both knock-out (KO) and knock-in (KI) mutations within the spider's offspring. The KO of the gene sine oculis causes total eye loss, confirming the role of the gene in the development of all spider eyes. The KI of a monomeric red fluorescent protein (mRFP-KI) within a spider silk gene encoding one compound of the major ampullate silk of the spider Parasteatoda tepidariorum yields red fluorescent silk fibers. This finding demonstrates the feasibility of functionalizing silk proteins in spiders using CRISPR-based gene editing without influencing silk assembly. Our study expands the application of CRISPR to spiders and provides insights in the fields of developmental genetics as well as material sciences.
Collapse
Affiliation(s)
- Edgardo Santiago-Rivera
- Department of Biomaterials, University of Bayreuth, Prof.-Rüdiger-Borman Strasse 1, 95448, Bayreuth, Germany
| | - Thomas Scheibel
- Department of Biomaterials, University of Bayreuth, Prof.-Rüdiger-Borman Strasse 1, 95448, Bayreuth, Germany
- Bayreuth Zentrum für Kolloide und Grenzflächen, Universität Bayreuth, 95440, Bayreuth, Germany
- Bayreuth Zentrum für Molekulare Biowissenshaften, Universität Bayreuth, 95440, Bayreuth, Germany
- Bayreuther Materialzentrum, Universität Bayreuth, 95440, Bayreuth, Germany
- Bayerisches Polymerinstitut, University of Bayreuth, 95440, Bayreuth, Germany
| |
Collapse
|
61
|
Almasoudi HH. Therapeutic promise of CRISPR-Cas9 gene editing in sickle cell disease and β-thalassemia: A current review. Curr Res Transl Med 2025; 73:103513. [PMID: 40252393 DOI: 10.1016/j.retram.2025.103513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 03/15/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
Sickle cell disease (SCD and β-thalassemia (BT) affects millions of people worldwide. In addition, around 500,000 infants are born with SCD and 60,000 people are diagnosed with BT every year. Mutations in the hemoglobin subunit beta (HBB) gene are responsible for causing both BT and SCD. Indeed, the diversity of potential mutations in the HBB gene elucidates the diversity in clinical severity observed in individuals with BT and related morbidities. On the other hand, SCD takes place because of the alteration in a single amino acid at position 6 in the beta-globin chain, where a base substitution occurs from glutamic acid to valine, which eventually results in abnormal sickle hemoglobin. Conventional therapies for BT and SCD including pharmaceutical drugs and blood transfusion might temporarily improve the clinical severity of these diseases, however these therapies cannot cure the diseases. CRISPR-Cas9 (CC9) is revolutionizing genome engineering, offering promising therapeutic avenues for genetic diseases. Therefore, CC9-mediated gene therapy provides great hope in the treatment of both BT and SCD. CC9-mediated gene therapy has already demonstrated its effectiveness in correcting both SCD and BT-causing mutations. Moreover, CC9-mediated gene editing was found to be effective in reactivating the expression of hemoglobin F (HbF) and regulating LRF and BCL11A. A number of clinical trials with CC9 gene-edited therapies are being carried out to elucidate their potential in treating BT and SCD. Genetics and pathophysiological mechanisms of SCD and BT, the mechanism of CC9-mediated gene editing, and common delivery methods of the CC9 system have been discussed in this review. Moreover, an in-depth discussion on applications and the current status of CC9-mediated gene editing in SCD and BT along with current challenges and future perspectives have been provided.
Collapse
Affiliation(s)
- Hassan H Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia.
| |
Collapse
|
62
|
Nam H, Han J, Yu J, Cho C, Kim D, Kim Y, Kim M, Kim J, Jo D, Bae S. Autophagy induction enhances homologous recombination-associated CRISPR-Cas9 gene editing. Nucleic Acids Res 2025; 53:gkaf258. [PMID: 40239991 PMCID: PMC11997770 DOI: 10.1093/nar/gkaf258] [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: 07/25/2024] [Revised: 02/24/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 (CRISPR-associated protein 9)-based gene editing via homologous recombination (HR) enables precise gene correction and insertion. However, its low efficiency poses a challenge due to the predominance of nonhomologous end-joining during DNA repair processes. Although numerous efforts have been made to boost HR efficiency, there remains a critical need to devise a novel method that can be universally applied across cell types and in vivo animals, which could ultimately facilitate therapeutic treatments. This study demonstrated that autophagy induction using different protocols, including nutrient deprivation or chemical treatment, significantly improved HR-associated gene editing at diverse genomic loci in mammalian cells. Notably, interacting cofactor proteins that bind to Cas9 under the autophagic condition have been identified, and autophagy induction could also enhance in vivo HR-associated gene editing in mice. These findings pave the way for effective gene correction or insertion for in vivo therapeutic treatments.
Collapse
Affiliation(s)
- Hye Jin Nam
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jun Hee Han
- Department of Chemistry, Hanyang University, Seoul 04673, Republic of Korea
| | - Jihyeon Yu
- Medical Research Center of Genomic Medicine Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Chang Sik Cho
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Dongha Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Young Eun Kim
- Center for Bioanalysis, Division of Chemical and Medical Metrology, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - Min Ji Kim
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jeong Hun Kim
- Fight Against Angiogenesis-Related Blindness (FARB) Laboratory, Biomedical Research Institute, Seoul National University, Seoul 03080, Republic of Korea
- Global Excellence Center for Gene & Cell Therapy (GEC-GCT), Seoul National University Hospital, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Dong Hyun Jo
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sangsu Bae
- Medical Research Center of Genomic Medicine Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
63
|
Salaudeen AL, Mateyko N, de Boer CG. RAPID-DASH: Single-Day Assembly of Guide RNA Arrays for Multiplexed CRISPR-Cas9 Applications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.09.648054. [PMID: 40291718 PMCID: PMC12027327 DOI: 10.1101/2025.04.09.648054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Guide RNA (gRNA) arrays can enable targeting multiple genomic loci simultaneously using CRISPR-Cas9. In this study, we present a streamlined and efficient method to rapidly construct gRNA arrays with up to 10 gRNA units in a single day. We demonstrate that gRNA arrays maintain robust functional activity across all positions, and can incorporate libraries of gRNAs, combining scalability and multiplexing. Our approach will streamline combinatorial perturbation research by enabling the economical and rapid construction, testing, and iteration of gRNA arrays.
Collapse
|
64
|
Bono H. Recent Advances in Genome Editing and Bioinformatics: Addressing Challenges in Genome Editing Implementation and Genome Sequencing. Int J Mol Sci 2025; 26:3442. [PMID: 40244417 PMCID: PMC11989416 DOI: 10.3390/ijms26073442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/30/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Genome-editing technology has advanced significantly since the 2020 Nobel Prize in Chemistry was awarded for the development of clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9). While CRISPR-Cas9 has become widely used in academic research, its social implementation has lagged due to unresolved patent disputes and slower progress in gene function analysis. To address this, new approaches bypassing direct gene function analysis are needed, with bioinformatics and next-generation sequencing (NGS) playing crucial roles. NGS is essential for sequencing the genome of target species, but challenges such as data quality, genome heterogeneity, ploidy, and small individual sizes persist. Despite these issues, advancements in sequencing technologies, like PacBio high-fidelity (HiFi) long reads and high-throughput chromosome conformation capture (Hi-C), have improved genome sequencing. Bioinformatics contributes to genome editing through off-target prediction and target gene selection, both of which require accurate genome sequence information. In this review, I will give updates on the development of genome editing and bioinformatics technologies with a focus on the rapid progress in genome sequencing.
Collapse
Affiliation(s)
- Hidemasa Bono
- Graduate School of Integrated Sciences for Life, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima 739-0046, Japan; ; Tel.: +81-82-424-4013
- Department of Biological Science, School of Science, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima 739-0046, Japan
- Genome Editing Innovation Center, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima 739-0046, Japan
| |
Collapse
|
65
|
Khan MS, Qureshi N, Khan R, Son YO, Maqbool T. CRISPR/Cas9-Based therapeutics as a promising strategy for management of Alzheimer's disease: progress and prospects. Front Cell Neurosci 2025; 19:1578138. [PMID: 40260080 PMCID: PMC12009953 DOI: 10.3389/fncel.2025.1578138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 03/20/2025] [Indexed: 04/23/2025] Open
Abstract
CRISPR/Cas9 technology has revolutionized genetic and biomedical research in recent years. It enables editing and modulation of gene function with an unparalleled precision and effectiveness. Among the various applications and prospects of this technology, the opportunities it offers in unraveling the molecular underpinnings of a myriad of central nervous system diseases, including neurodegenerative disorders, psychiatric conditions, and developmental abnormalities, are unprecedented. In this review, we highlight the applications of CRISPR/Cas9-based therapeutics as a promising strategy for management of Alzheimer's disease and transformative impact of this technology on AD research. Further, we emphasize the role of CRISPR/Cas9 in generating accurate AD models for identification of novel therapeutic targets, besides the role of CRISPR-based therapies aimed at correcting AD-associated mutations and modulating the neurodegenerative processes. Furthermore, various delivery systems are reviewed and potential of the non-viral nanotechnology-based carriers for overcoming the critical limitations of effective delivery systems for CRISPR/Cas9 is discussed. Overall, this review highlights the promise and prospects of CRISPR/Cas9 technology for unraveling the intricate molecular processes underlying the development of AD, discusses its limitations, ethical concerns and several challenges including efficient delivery across the BBB, ensuring specificity, avoiding off-target effects. This article can be helpful in better understanding the applications of CRISPR/Cas9 based therapeutic approaches and the way forward utilizing enormous potential of this technology in targeted, gene-specific treatments that could change the trajectory of this debilitating and incurable illness.
Collapse
Affiliation(s)
- Mohamad Sultan Khan
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| | - Nousheen Qureshi
- Department of Higher Education, Government of Jammu and Kashmir, Srinagar, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Mohali, Punjab, India
| | - Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences and Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, Republic of Korea
| | - Tariq Maqbool
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| |
Collapse
|
66
|
He Q, Wang Y, Tan Z, Zhang X, Yu C, Jiang X. Mapping the therapeutic landscape of CRISPR-Cas9 for combating age-related diseases. Front Genome Ed 2025; 7:1558432. [PMID: 40255230 PMCID: PMC12006052 DOI: 10.3389/fgeed.2025.1558432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/19/2025] [Indexed: 04/22/2025] Open
Abstract
CRISPR-Cas9 (clustered regularly interspaced short palindromic repeats-associated protein 9) has emerged as a transformative genome-editing tool with significant therapeutic potential for age-related diseases, including Alzheimer's disease, Parkinson's disease, cardiovascular disorders, and osteoporosis. This study presents a bibliometric analysis of CRISPR-Cas9 research in age-related diseases, identifying key contributors, major research hotspots, and critical technological advancements. While promising applications have been demonstrated in gene repair, functional regulation, and molecular interventions, significant barriers persist, including off-target effects, low delivery efficiency, and limited editing in non-dividing cells. Ethical concerns over germline editing and gaps in long-term safety data further complicate clinical translation. Future directions emphasize the development of high-precision Cas9 variants, homology-directed repair-independent tools, and efficient delivery systems, alongside the establishment of international regulatory frameworks and multicenter clinical trials. These efforts are essential to fully realize the potential of CRISPR-Cas9 in addressing the global health challenges of aging.
Collapse
Affiliation(s)
- Qiyu He
- Department of Urology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yida Wang
- Key Laboratory of BioResource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu, Sichuan, China
| | - Zhimin Tan
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xian Zhang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Chao Yu
- Department of Anesthesiology, West China Second Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Xiaoqin Jiang
- Department of Anesthesiology, West China Second Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Anesthesiology, Chengdu Hi-Tech Zone Hospital for Women and Children, Chengdu, China
| |
Collapse
|
67
|
Zhang Y, Lin S, Yu L, Lin X, Qu S, Ye Q, Yu M, Chen W, Wu W. Gene therapy shines light on congenital stationary night blindness for future cures. J Transl Med 2025; 23:392. [PMID: 40181393 PMCID: PMC11969737 DOI: 10.1186/s12967-025-06392-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/17/2025] [Indexed: 04/05/2025] Open
Abstract
Congenital Stationary Night Blindness (CSNB) is a non-progressive hereditary eye disease that primarily affects the retinal signal processing, resulting in significantly reduced vision under low-light conditions. CSNB encompasses various subtypes, each with distinct genetic patterns and pathogenic genes. Over the past few decades, gene therapy for retinal genetic disorders has made substantial progress; however, effective clinical therapies for CSNB are yet to be discovered. With the continuous advancement of gene-therapy tools, there is potential for these methods to become effective treatments for CSNB. Nonetheless, challenges remain in the treatment of CSNB, including issues related to delivery vectors, therapeutic efficacy, and possible side effects. This article reviews the clinical diagnosis, pathogenesis, and associated mutated genes of CSNB, discusses existing animal models, and explores the application of gene therapy technologies in retinal genetic disorders, as well as the current state of research on gene therapy for CSNB.
Collapse
Affiliation(s)
- Yi Zhang
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Siqi Lin
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Lingqi Yu
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Xiang Lin
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
- Department of Biomedical Engineering, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Shuai Qu
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
- Department of Biomedical Engineering, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Qingyang Ye
- Hangzhou Bipolar Biotechnology Co., Ltd., Hangzhou, 311199, China
| | - Mengting Yu
- Department of Ophthalmology, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350028, China
| | - Wenfeng Chen
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China.
- Department of Biomedical Engineering, College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China.
| | - Wenjie Wu
- Department of Ophthalmology, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350028, China.
| |
Collapse
|
68
|
Tachida Y, Manian KV, Butcher R, Levy JM, Pendse N, Hennessey E, Liu DR, Pierce EA, Liu Q, Comander J. Systematic empirical evaluation of individual base editing targets: Validating therapeutic targets in USH2A and comparison of methods. Mol Ther 2025; 33:1466-1484. [PMID: 39881543 PMCID: PMC11997516 DOI: 10.1016/j.ymthe.2025.01.042] [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: 08/23/2024] [Revised: 11/22/2024] [Accepted: 01/24/2025] [Indexed: 01/31/2025] Open
Abstract
Base editing shows promise for the correction of human mutations at a higher efficiency than other repair methods and is especially attractive for mutations in large genes that are not amenable to gene augmentation therapy. Here, we demonstrate a comprehensive workflow for in vitro screening of potential therapeutic base editing targets for the USH2A gene and empirically validate the efficiency of adenine and cytosine base editor/guide combinations for correcting 35 USH2A mutations. Editing efficiency and bystander edits are compared between different target templates (plasmids vs. transgenes) and assays (next-generation sequencing vs. Sanger), as well as comparisons between unbiased empirical results and computational predictions. Based on these observations, practical assay recommendations are discussed. Finally, a humanized knockin mouse model was created with the best-performing target, the nonsense mutation c.11864G>A p.(Trp3955∗). Split-intein AAV9 delivery of editing reagents resulted in the restoration of USH2A protein and a correction rate of 65% ± 3% at the mutant base pair and of 52% ± 3% excluding bystander amino acid changes. This efficiency is higher than that seen in a retinal gene editing program testing in a clinical trial. These results demonstrate the effectiveness of this overall strategy to identify and test base editing reagents with the potential for human therapeutic applications.
Collapse
Affiliation(s)
- Yuki Tachida
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Kannan V Manian
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Rossano Butcher
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Jonathan M Levy
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02142, USA
| | - Nachiket Pendse
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Erin Hennessey
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02142, USA
| | - Eric A Pierce
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Qin Liu
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Jason Comander
- Ocular Genomics Institute, Berman-Gund Laboratory for the Study of Retinal Degenerations, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
69
|
Ahn SJ. Exploring neuropharmacokinetics: mechanisms, models, and clinical implications. ENCEPHALITIS 2025; 5:36-52. [PMID: 40194559 PMCID: PMC12042691 DOI: 10.47936/encephalitis.2024.00080] [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: 08/21/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 04/09/2025] Open
Abstract
Neuropharmacokinetics is an emerging field dedicated to understanding the pharmacokinetics of drugs within the central nervous system (CNS), with a particular emphasis on overcoming the challenges posed by the blood-brain barrier. This paper reviews the latest advancements in drug delivery strategies, including nanoparticle-based systems, receptor-mediated transcytosis, and efflux transporter inhibition, which have been designed to enhance drug penetration into the brain. Additionally, the use of advanced imaging techniques such as positron emission tomography, functional magnetic resonance imaging, and magnetic resonance imaging with contrast agents has provided critical insights into drug distribution, receptor occupancy, and the functional impact of therapeutic agents within the CNS. These innovations not only enhance our understanding of CNS drug action but also pave the way for more effective treatments for neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Seon-Jae Ahn
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
- Center for Hospital Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
70
|
Pirali A, Jafarpour F, Hajian M, Hosseini Moghaddam SH, Moradi R, Tanhaie-Vash N, Rahimi Andani M, Izadi T, Shiralian-Esfahani H, Safaeinejad Z, Kues W, Nasr-Esfahani MH, Eghbalsaied S. Editing the CYP19 Gene in Goat Embryos Using CRISPR/Cas9 and Somatic Cell Nuclear Transfer Techniques. Cell Reprogram 2025; 27:86-93. [PMID: 40126138 DOI: 10.1089/cell.2024.0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025] Open
Abstract
The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR/Cas9) system is revolutionizing genome engineering and is expected to bring significant advancements in livestock traits, including the treatment of genetic diseases. This study focuses on CRISPR/Cas9-mediated modifications of the CYP19 gene, which encodes aromatase, an enzyme crucial for converting testosterone to estrogen and essential for steroid metabolism. Guide RNAs (gRNAs) were designed to target the CYP19 gene and cloned into the pX459 vector. The recombinant plasmid was then electrotransfected into fibroblast cells from a Lori-Bakhtiari buck, and these transfected cells were used for embryo production via somatic cell nuclear transfer (SCNT). The cloned embryos were evaluated for their progression through embryonic stages, showing no significant difference in blastocyst development between knock-out and unedited groups. The knockout efficiency was 78.4% in cells and 68.9% in goat blastocysts, demonstrating the successful depletion of CYP19. We successfully achieved a high rate of CYP19 gene-edited embryos through the combined application of cell electrotransfection and SCNT technologies, while maintaining the normal developmental rate of the embryos. These embryos can be used for transfer to generate knock-out goats, providing a foundation for further studies on CYP19's role in male fertility and production traits.
Collapse
Affiliation(s)
- Ahmad Pirali
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- Faculty of Agricultural Sciences, Department of Animal Science, University of Guilan, Rasht, Iran
| | - Farnoosh Jafarpour
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mehdi Hajian
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | | | - Reza Moradi
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Nima Tanhaie-Vash
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohsen Rahimi Andani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Tayebeh Izadi
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Hanieh Shiralian-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Zahra Safaeinejad
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Wilfried Kues
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Neustadt Rbge, Germany
| | - Mohammad-Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Shahin Eghbalsaied
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- Department of Animal Science, Isfahan (Khorasgan) Branch, Islamic Azad University, Tehran, Iran
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
71
|
Han G, Hu K, Luo T, Wang W, Zhang D, Ouyang L, Liu X, Liu J, Wu Y, Liang J, Ling J, Chen Y, Xuan R, Zhang J, Yu P. Research progress of non-coding RNA regulating the role of PANoptosis in diabetes mellitus and its complications. Apoptosis 2025; 30:516-536. [PMID: 39755822 DOI: 10.1007/s10495-024-02066-w] [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] [Accepted: 12/20/2024] [Indexed: 01/06/2025]
Abstract
Diabetes is a chronic metabolic disease that is endemic worldwide and is characterized by persistent hyperglycemia accompanied by multiple severe complications, including cardiovascular disease, kidney dysfunction, neuropathy, and retinopathy. The pathogenesis of diabetes mellitus and its complications is multifactorial, involving various molecular and cellular pathways. In recent years, research has indicated that mechanisms of cell death play a significant role in the advancement of diabetes and its complications. PANoptosis is a complex phenomenon caused by three cell death pathways: programmed apoptosis, necroptosis and pyroptosis. The contribution of PANoptosis to diabetes and its complications remains incompletely understood. Non-coding RNA, an important molecule in gene expression regulation, has shown significant regulatory functions in a variety of diseases. This paper reviews the underlying mechanisms of diverse types of non-coding RNAs (including lncRNA, miRNA and circRNA) in regulating PANoptosis and their specific contributions in diabetes, aiming to explore how non-coding RNAs influence PANoptosis and their effects in diabetes.
Collapse
Affiliation(s)
- Guangyu Han
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Kaibo Hu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Tianfeng Luo
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Wenting Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Hainan Medical University, Haikou, 571199, China
| | - Deju Zhang
- Ood and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Liu Ouyang
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street SE, Atlanta, GA, 30303, USA
| | - Xiao Liu
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jianqi Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Rui Xuan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
72
|
Lv J, Fan Q, Zhang Y, Zhou X, Yu P, Yu X, Xin C, Hong J, Cheng Y. A Serum Resistant Polymer with Exceptional Endosomal Escape and mRNA Delivery Efficacy for CRISPR Gene Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413006. [PMID: 39921871 PMCID: PMC11967772 DOI: 10.1002/advs.202413006] [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: 10/17/2024] [Revised: 12/16/2024] [Indexed: 02/10/2025]
Abstract
Nanoparticle-based mRNA delivery offers a versatile platform for innovative therapies. However, most of the current delivery systems are limited by poor serum tolerance, suboptimal endosomal escape and mRNA delivery efficacy. Herein, a highly efficient mRNA-delivering material is identified from a library of fluoropolymers. The lead material FD17 shows exceptional serum stability and endosomal escape, enabling efficient mRNA delivery into various cell types, surpassing commercial mRNA delivery reagents such as Lipofectamine 3000. The formed mRNA nanoparticles adsorb abundant serum albumin on the surface, which facilitates cellular uptake via scavenger receptor-mediated endocytosis. FD17 enables the delivery of mRNAs encoding CRE, Cas9, and base editor hyCBE for efficient genome editing. The material mediates CRISPR/Cas9 gene therapy via intraocular injection effectively down-regulates vascular endothelial growth factor A in retinal pigment epithelial cells of mice, yielding promising therapeutic responses against laser-induced choroidal neovascularization. The discovered material in this study shows great promise for the development of mRNA therapeutics to combat a wide range of diseases.
Collapse
Affiliation(s)
- Jia Lv
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal UniversityShanghai200241China
| | - Qianqian Fan
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal UniversityShanghai200241China
- Department of General SurgeryCenter for Metabolism ResearchThe Fourth Affiliated Hospital of Zhejiang University School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
| | - Yirou Zhang
- Department of Ophthalmology and Vision ScienceEye, Ear, Nose, and Throat HospitalFudan UniversityShanghai200030China
| | - Xujiao Zhou
- Department of Ophthalmology and Vision ScienceEye, Ear, Nose, and Throat HospitalFudan UniversityShanghai200030China
| | - Panting Yu
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal UniversityShanghai200241China
| | - Xin Yu
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal UniversityShanghai200241China
| | - Changchang Xin
- Department of Ophthalmology and Vision ScienceEye, Ear, Nose, and Throat HospitalFudan UniversityShanghai200030China
| | - Jiaxu Hong
- Department of Ophthalmology and Vision ScienceEye, Ear, Nose, and Throat HospitalFudan UniversityShanghai200030China
- Department of OphthalmologyEye & ENT HospitalState Key Laboratory of Molecular Engineering of PolymersFudan UniversityShanghai200031China
- NHC Key laboratory of Myopia and Related Eye DiseasesShanghai200031China
- Shanghai Engineering Research Center of Synthetic ImmunologyShanghai200032China
- Department of OphthalmologyChildren's Hospital of Fudan UniversityNational Pediatric Medical Center of ChinaShanghai200031China
| | - Yiyun Cheng
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory BiologySchool of Life SciencesEast China Normal UniversityShanghai200241China
| |
Collapse
|
73
|
Hong H, Han H, Wang L, Cao W, Hu M, Li J, Wang J, Yang Y, Xu X, Li G, Zhang Z, Zhang C, Xu M, Wang H, Wang Q, Yuan Y. ABCF1-K430-Lactylation promotes HCC malignant progression via transcriptional activation of HIF1 signaling pathway. Cell Death Differ 2025; 32:613-631. [PMID: 39753865 PMCID: PMC11982231 DOI: 10.1038/s41418-024-01436-w] [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: 05/28/2024] [Revised: 12/08/2024] [Accepted: 12/16/2024] [Indexed: 01/12/2025] Open
Abstract
Lysine lactylation plays critical roles in various diseases, including cancer. Our previous study showed that lactylation of non-histone ABCF1 may be involved in hepatocellular carcinoma (HCC) progression. In this study, we evaluated the prognostic value of ABCF1-K430la in HCC using immunohistochemical staining and performed amino acid point mutations, multi-omics crossover, and biochemical experiments to investigate its biological role and underlying mechanism. Additionally, we performed molecular docking on lactylation sites. ABCF1-K430la was highly expressed in HCC tissues and correlated with poor patient prognosis. Functionally, ABCF1-K430la promoted HCC growth and lung metastasis. Mechanistically, upon lactylation, E2 ubiquitin ligase activity of ABCF1 remained unaffected, and ABCF1 entered the nucleus, bound to the KDM3A promoter to upregulate its expression, and activated the KDM3A-H3K9me2-HIF1A axis, challenging the notion that ABCF1 functions exclusively in cytoplasmic protein translation. Notably, we discovered the existence of a lactate-ABCF1(430Kla)-HIF1A-lactate in HCC. A small-molecule drug screen targeting ABCF1-K430la revealed that tubuloside A inhibits ABCF1-K430la and suppresses HCC development. These findings demonstrate that elevated ABCF1-K430la expression promotes HCC development, suggesting it as a potential prognostic biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Han Hong
- Department of Hepato-Pancreato-Biliary Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hexu Han
- Department of Gastroenterology of the affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Lei Wang
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Wen Cao
- Department of Liver Disease of the affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Minjie Hu
- Department of Hepatobiliary Surgery of the affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Jindong Li
- Department of Pharmacy of the affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Jiawei Wang
- Department of Hepatobiliary Surgery of the affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Yijin Yang
- Department of Oncology, Suzhou Kowloon Hospital, Shanghai Jiao Tong University, School of Medicine, Suzhou, China
| | - XiaoYong Xu
- Department of Hepato-Pancreato-Biliary Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Gaochao Li
- Department of Hepato-Pancreato-Biliary Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Zixiang Zhang
- Department of General Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Changhe Zhang
- Department of Hepatobiliary Surgery of the affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China.
| | - Minhui Xu
- Department of Hepato-Pancreato-Biliary Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
| | - Honggang Wang
- Department of Gastrointestinal Surgery of the affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China.
| | - Qiang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; Innovative Institute of Tumor Immunity and Medicine (ITIM); Anhui Provincial Innovation Institute for Pharmaceutical Basic Research; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, China.
| | - Yin Yuan
- Department of Hepatobiliary Surgery of the affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China.
- Department of Clinical research center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China.
| |
Collapse
|
74
|
Palomino SM, Gabriel KA, Mwirigi JM, Cervantes A, Horton P, Funk G, Moutal A, Martin LF, Khanna R, Price TJ, Patwardhan A. Genetic editing of primary human dorsal root ganglion neurons using CRISPR-Cas9. Sci Rep 2025; 15:11116. [PMID: 40169710 PMCID: PMC11961745 DOI: 10.1038/s41598-025-91153-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/18/2025] [Indexed: 04/03/2025] Open
Abstract
CRISPR-Cas9 is now the leading method for genome editing and is advancing for the treatment of human disease. CRIPSR has promise in treating neurological diseases, but traditional viral-vector-delivery approaches have neurotoxicity limiting their use. Here we describe a simple method for non-viral transfection of primary human DRG (hDRG) neurons for CRISPR-Cas9 editing. We edited TRPV1, NTSR2, and CACNA1E using a lipofection method with CRISPR-Cas9 plasmids containing reporter tags (GFP or mCherry). Transfection was successfully demonstrated by the expression of the reporters two days post-administration. CRISPR-Cas9 editing was confirmed at the genome level with a T7-endonuclease-I assay; protein level with immunocytochemistry and Western blot; and functional level through capsaicin-induced Ca2+ accumulation in a high-throughput compatible fluorescent imaging plate reader (FLIPR) system. This work establishes a reliable, target specific, non-viral CRISPR-Cas9-mediated genetic editing in primary human neurons with potential for future clinical application for sensory diseases.
Collapse
Affiliation(s)
- Seph M Palomino
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, 6202 Harry Hines Blvd., 9th Floor, Dallas, 75235, TX, USA
| | - Katherin A Gabriel
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Juliet M Mwirigi
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Anna Cervantes
- Southwest Transplant Alliance, Manderville Ln, Dallas, TX, 8190, 75231, USA
| | - Peter Horton
- Southwest Transplant Alliance, Manderville Ln, Dallas, TX, 8190, 75231, USA
| | - Geoffrey Funk
- Southwest Transplant Alliance, Manderville Ln, Dallas, TX, 8190, 75231, USA
| | - Aubin Moutal
- Department of Pharmacology and Physiology, Saint Louis University, 1402 S. Grand Blvd, St. Louis, Mo, 63104, USA
| | - Laurent F Martin
- Department of Pharmacology, University of Arizona, 1501 N Campbell Ave, Tucson, AZ, 85721, USA
| | - Rajesh Khanna
- Department of Pharmacology and Therapeutics, University of Florida, 1200 Newell Drive, Gainesville, FL, ARB R5-234, 32610-0267, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA.
| | - Amol Patwardhan
- Department of Anesthesiology and Pain Management, University of Texas Southwestern Medical Center, 6202 Harry Hines Blvd., 9th Floor, Dallas, 75235, TX, USA.
| |
Collapse
|
75
|
Kwon YW. Illuminating the Protective Role of UPF1 in Myocardial Ischemia/Reperfusion Injury. Korean Circ J 2025; 55:322-324. [PMID: 40205999 PMCID: PMC12046307 DOI: 10.4070/kcj.2025.0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/05/2025] [Accepted: 02/09/2025] [Indexed: 04/11/2025] Open
Affiliation(s)
- Yoo-Wook Kwon
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Department of Medicine, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
76
|
Zhang J, Zhou Y, Qiao J, Liu Y. Recent advances in spatiotemporal control of the CRISPR/Cas9 system. Colloids Surf B Biointerfaces 2025; 248:114474. [PMID: 39732069 DOI: 10.1016/j.colsurfb.2024.114474] [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: 10/31/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
The CRISPR/Cas9 gene-editing technology, derived from the adaptive immune mechanisms of bacteria, has demonstrated remarkable advantages in fields such as gene function research and the treatment of genetic diseases due to its simplicity in design, precise targeting, and ease of use. Despite challenges such as off-target effects and cytotoxicity, effective spatiotemporal control strategies have been achieved for the CRISPR/Cas9 system through precise regulation of Cas9 protein activity as well as engineering of guide RNAs (gRNAs). This review provides a comprehensive analysis of the core components and functional mechanisms underlying the CRISPR/Cas9 system, highlights recent advancements in spatiotemporal control strategies, and discusses future directions for development.
Collapse
Affiliation(s)
- Junqi Zhang
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, Hubei 430023, China; School of Life Sciences, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, Hubei 430042, China
| | - Yuzi Zhou
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, Hubei 430023, China
| | - Jie Qiao
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, Hubei 430023, China.
| | - Yi Liu
- School of Life Sciences, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, Hubei 430042, China.
| |
Collapse
|
77
|
Gu B, Li M, Li D, Huang K. CRISPR-Cas9 Targeting PCSK9: A Promising Therapeutic Approach for Atherosclerosis. J Cardiovasc Transl Res 2025; 18:424-441. [PMID: 39804565 DOI: 10.1007/s12265-024-10587-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 12/28/2024] [Indexed: 05/01/2025]
Abstract
CRISPR-Cas9 gene editing technology, as an innovative biomedical tool, holds significant potential in the prevention and treatment of atherosclerosis. By precisely editing key genes such as PCSK9, CRISPR-Cas9 offers the possibility of long-term regulation of low-density lipoprotein cholesterol (LDL-C), which may reduce the risk of cardiovascular diseases. Early clinical studies of gene editing therapies like VERVE-101 have yielded encouraging results, highlighting both the feasibility and potential efficacy of this technology. However, clinical applications still face challenges such as off-target effects, immunogenicity, and long-term safety. Future research should focus on enhancing the specificity and efficiency of gene editing, optimizing delivery systems, and improving personalized treatment strategies. Additionally, the establishment of ethical and legal regulatory frameworks will be critical for the safe adoption of this technology. With the continued advancement of gene editing technology, CRISPR-Cas9 may become an important tool for treating atherosclerosis and other complex diseases.
Collapse
Affiliation(s)
- Bin Gu
- Department of Cardiology, Affiliated Hospital of Southwest Medical University, No.1 Section 1, Xiang Lin Road, Longmatan District, Luzhou, Sichuan, 646000, China
| | - Min Li
- Department of Cardiology, Neijiang Dongxing District People's Hospital, Neijiang, Sichuan, 641300, China
| | - Dan Li
- Department of Cardiology, Neijiang Dongxing District People's Hospital, Neijiang, Sichuan, 641300, China
| | - Kaisen Huang
- Department of Cardiology, Affiliated Hospital of Southwest Medical University, No.1 Section 1, Xiang Lin Road, Longmatan District, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
78
|
Zheng Y, Zou Q, Li J, Yang Y. CRISPR-MFH: A Lightweight Hybrid Deep Learning Framework with Multi-Feature Encoding for Improved CRISPR-Cas9 Off-Target Prediction. Genes (Basel) 2025; 16:387. [PMID: 40282347 PMCID: PMC12026807 DOI: 10.3390/genes16040387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND The CRISPR-Cas9 system has emerged as one of the most promising gene-editing technologies in biology. However, off-target effects remain a significant challenge. While recent advances in deep learning have led to the development of models for off-target prediction, these models often fail to fully leverage sequence pair information. Furthermore, as the models' parameter sizes increase, so do their complexities, limiting their practical applicability. METHODS In this study, we introduce a novel multi-feature independent encoding method, which encodes the gRNA-DNA sequence pair into three distinct feature matrices to minimize information loss. Additionally, we propose a lightweight hybrid deep learning framework, CRISPR-MFH, that integrates multi-scale separable convolutions and hybrid attention mechanisms for efficient and accurate off-target prediction. RESULTS Extensive experiments across multiple benchmark datasets demonstrate that the proposed encoding method effectively captures critical features and that CRISPR-MFH outperforms or matches state-of-the-art models with significantly fewer parameters across multiple evaluation metrics. CONCLUSIONS This study offers a novel perspective for advancing deep learning technology in the realm of CRISPR-Cas9 off-target detection.
Collapse
Affiliation(s)
- Yanyi Zheng
- College of Landscape Architecture, Beijing Forestry University, Beijing 100083, China;
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China;
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China
| | - Jian Li
- School of Mathematics and Computer Science, Zhejiang A&F University, Hangzhou 311300, China
| | - Yanpeng Yang
- School of Mathematics and Computer Science, Zhejiang A&F University, Hangzhou 311300, China
| |
Collapse
|
79
|
Kordana N, Johnson A, Quinn K, Obar JJ, Cramer RA. Recent developments in Aspergillus fumigatus research: diversity, drugs, and disease. Microbiol Mol Biol Rev 2025; 89:e0001123. [PMID: 39927770 PMCID: PMC11948498 DOI: 10.1128/mmbr.00011-23] [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] [Indexed: 02/11/2025] Open
Abstract
SUMMARYAdvances in modern medical therapies for many previously intractable human diseases have improved patient outcomes. However, successful disease treatment outcomes are often prevented due to invasive fungal infections caused by the environmental mold Aspergillus fumigatus. As contemporary antifungal therapies have not experienced the same robust advances as other medical therapies, defining mechanisms of A. fumigatus disease initiation and progression remains a critical research priority. To this end, the World Health Organization recently identified A. fumigatus as a research priority human fungal pathogen and the Centers for Disease Control has highlighted the emergence of triazole-resistant A. fumigatus isolates. The expansion in the diversity of host populations susceptible to aspergillosis and the complex and dynamic A. fumigatus genotypic and phenotypic diversity call for a reinvigorated assessment of aspergillosis pathobiological and drug-susceptibility mechanisms. Here, we summarize recent advancements in the field and discuss challenges in our understanding of A. fumigatus heterogeneity and its pathogenesis in diverse host populations.
Collapse
Affiliation(s)
- Nicole Kordana
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Angus Johnson
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Katherine Quinn
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Joshua J. Obar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Robert A. Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
80
|
Ma E, Chen K, Shi H, Wasko KM, Esain-Garcia I, Trinidad MI, Zhou K, Ye J, Doudna JA. Directed evolution expands CRISPR-Cas12a genome editing capacity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645588. [PMID: 40196639 PMCID: PMC11974813 DOI: 10.1101/2025.03.26.645588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
CRISPR-Cas12a enzymes are versatile RNA-guided genome-editing tools with applications encompassing viral diagnosis, agriculture and human therapeutics. However, their dependence on a 5'-TTTV-3' protospacer-adjacent motif (PAM) next to DNA target sequences restricts Cas12a's gene targeting capability to only ∼1% of a typical genome. To mitigate this constraint, we used a bacterial-based directed evolution assay combined with rational engineering to identify variants of Lachnospiraceae bacterium Cas12a (LbCas12a) with expanded PAM recognition. The resulting Cas12a variants use a range of non-canonical PAMs while retaining recognition of the canonical 5'-TTTV-3' PAM. In particular, biochemical and cell-based assays show that the variant Flex-Cas12a utilizes 5'-NYHV-3' PAMs that expand DNA recognition sites to ∼25% of the human genome. With enhanced targeting versatility, Flex-Cas12a unlocks access to previously inaccessible genomic loci, providing new opportunities for both therapeutic and agricultural genome engineering.
Collapse
|
81
|
Li T, Raja BR, Liao J, Zheng L, Yin F, Gan S, Sun X, Lyu G, Ma J. The characteristics, influence factors, and regulatory strategies of growth retardation in ruminants: a review. Front Vet Sci 2025; 12:1566427. [PMID: 40206254 PMCID: PMC11979133 DOI: 10.3389/fvets.2025.1566427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Growth retardation represents a main barrier to affect the productivity and efficiency of ruminants production, which is characterized by low growth rate, a disparity between skeletal and physiological maturation, gastrointestinal dysfunction and reduced reproductive performance. This review provides a concise overview of growth retardation in ruminants, and summarizes the key factors that influence their growth and development, including genetics, nutrition, microbiota and environment. Also, this review emphasizes the central role of nutritional management and gastrointestinal development, as well as the regulatory mechanisms involved in growth processes. In addition, recent advances in these aspects are discussed to form an integrative framework aimed at improving physiological function in ruminants. This review provides a comprehensive perspective for understanding the complex mechanism of growth retardation in ruminants, puts forward a theoretical basis for optimizing the production efficiency of ruminants industry and emphasizes the importance of multidisciplinary collaboration to provide a reference for advancing systematic research on growth and development of ruminants.
Collapse
Affiliation(s)
- Tao Li
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Bakhtawar Riaz Raja
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Jie Liao
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Longqing Zheng
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Fuquan Yin
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Shangquan Gan
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Xuemei Sun
- Xinjiang Taikun Group Co., Ltd., Changji, China
| | - Gang Lyu
- Xinjiang Taikun Group Co., Ltd., Changji, China
| | - Jian Ma
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
- Xinjiang Taikun Group Co., Ltd., Changji, China
| |
Collapse
|
82
|
Zhu Z, Shen J, Ho PCL, Hu Y, Ma Z, Wang L. Transforming cancer treatment: integrating patient-derived organoids and CRISPR screening for precision medicine. Front Pharmacol 2025; 16:1563198. [PMID: 40201690 PMCID: PMC11975957 DOI: 10.3389/fphar.2025.1563198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
The persistently high mortality rates associated with cancer underscore the imperative need for innovative, efficacious, and safer therapeutic agents, as well as a more nuanced understanding of tumor biology. Patient-derived organoids (PDOs) have emerged as innovative preclinical models with significant translational potential, capable of accurately recapitulating the structural, functional, and heterogeneous characteristics of primary tumors. When integrated with cutting-edge genomic tools such as CRISPR, PDOs provide a powerful platform for identifying cancer driver genes and novel therapeutic targets. This comprehensive review delves into recent advancements in CRISPR-mediated functional screens leveraging PDOs across diverse cancer types, highlighting their pivotal role in high-throughput functional genomics and tumor microenvironment (TME) modeling. Furthermore, this review highlights the synergistic potential of integrating PDOs with CRISPR screens in cancer immunotherapy, focusing on uncovering immune evasion mechanisms and improving the efficacy of immunotherapeutic approaches. Together, these cutting-edge technologies offer significant promise for advancing precision oncology.
Collapse
Affiliation(s)
- Ziyi Zhu
- The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, Hubei, China
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Jiayang Shen
- The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, Hubei, China
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Paul Chi-Lui Ho
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
| | - Ya Hu
- The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, Hubei, China
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Zhaowu Ma
- The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, Hubei, China
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, Singapore
| |
Collapse
|
83
|
Carman LE, Samulevich ML, Aneskievich BJ. Protocol Development for CRISPR/Cas9 Knockout of the Anti-inflammatory Protein TNIP1 in HaCaT Keratinocytes. Methods Mol Biol 2025. [PMID: 40106146 DOI: 10.1007/7651_2025_616] [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: 03/22/2025]
Abstract
Gene editing in cultured cells, including the intent of sequence disruption to achieve a functional knockout of the targeted gene, has been greatly facilitated with the advent of Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)/CRISPR-associated protein 9 (CRISPR/Cas9) technology. Primary cell strains and immortalized cell lines from diverse tissue types have been successfully targeted both for basic research examining the effects of loss of the correlating protein and for modeling select loss-of-function disorders. Such accomplishments have extended to cutaneous cells, especially epidermal keratinocytes given their key structural and functional role in barrier formation and surveillance of and response to surface events such as triggering and processing inflammatory responses. Here we describe disruption of the Tumor Necrosis factor-induced protein 3-Interacting Protein 1 (TNIP1) gene in human HaCaT keratinocytes to generate an ongoing loss of expression as a parallel system to transient knockdown we had previously achieved with siRNA transfection. The TNIP1 protein restricts cytoplasmic progression of inflammatory signals. We cover our CRISPR/Cas9 vector choice, enrichment of transfected cells via positive selection for puromycin resistance, their subsequent cloning, and gene disruption and expression analysis. We also emphasize prior keratinocyte-CRISPR/Cas9 literature as a springboard for other investigators and to illustrate the widespread relevance of such editing to the diverse, yet highly consequentially different, genes expressed in keratinocytes.
Collapse
Affiliation(s)
- Liam E Carman
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT, USA
| | - Michael L Samulevich
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT, USA
| | - Brian J Aneskievich
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
84
|
He X, Yan T, Song Z, Xiang L, Xiang J, Yang Y, Ren K, Bu J, Xu X, Li Z, Guo X, Lin B, Zhou Q, Lin G, Gu F. Correcting a patient-specific Rhodopsin mutation with adenine base editor in a mouse model. Mol Ther 2025:S1525-0016(25)00195-9. [PMID: 40119518 DOI: 10.1016/j.ymthe.2025.03.021] [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: 08/10/2024] [Revised: 01/05/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
Genome editing offers a great promise to treating human genetic diseases. To assess genome-editing-mediated therapeutic effects in vivo, an animal model is indispensable. The genomic disparities between mice and humans often impede the direct clinical application of genome-editing-mediated treatments using conventional mouse models. Thus, the generation of a mouse model with a humanized genomic segment containing a patient-specific mutation is highly sought after for translational research. In this study, we successfully developed a knockin mouse model for autosomal-dominant retinitis pigmentosa (adRP), designated as hT17M knockin, which incorporates a 75-nucleotide DNA segment with the T17M mutation (Rhodopsin-c.C50T; p.T17M). This model demonstrated significant reductions in electroretinogram amplitudes and exhibited disruptions in retinal structure. Subsequently, we administered an adeno-associated virus vectors carrying an adenine base editor (ABE) and a single-guide RNA specifically targeting the T17M mutation, achieving a peak correction rate of 39.7% at the RNA level and significantly improving retinal function in ABE-injected mice. These findings underscore that the hT17M knockin mouse model recapitulates the clinical features of adRP patients and exhibits therapeutic effects with ABE-mediated treatments. It offers a promising avenue for the development of gene-editing therapies for RP.
Collapse
Affiliation(s)
- Xiaoxue He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, Hunan 410081, China; School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Tong Yan
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Zongming Song
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University and People's Hospital of Henan University, Zhengzhou, Henan 450003, China
| | - Lue Xiang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Jiayang Xiang
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China
| | - Yeqin Yang
- School of Nursing, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Kaiqun Ren
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, Hunan 410081, China
| | - Jicheng Bu
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan 410006, China
| | - Xilin Xu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, Hunan 410081, China; Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan 410006, China
| | - Zhuo Li
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan 410006, China
| | - Xiaowei Guo
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, Hunan 410081, China
| | - Bin Lin
- School of Optometry, Hong Kong Polytechnic University, Hong Kong HJ502, China
| | - Qinghua Zhou
- Department of Anesthesiology, The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Ge Lin
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan 410006, China
| | - Feng Gu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, Hunan 410081, China; School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, State Key Laboratory and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, Wenzhou, Zhejiang 325027, China; Guangxiu Hospital Affiliated with Hunan Normal University (Hunan Guangxiu Hospital), Changsha, Hunan 410119, China.
| |
Collapse
|
85
|
Liu D, Cao D, Han R. Recent advances in therapeutic gene-editing technologies. Mol Ther 2025:S1525-0016(25)00200-X. [PMID: 40119516 DOI: 10.1016/j.ymthe.2025.03.026] [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: 12/13/2024] [Revised: 02/26/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
The advent of gene-editing technologies, particularly CRISPR-based systems, has revolutionized the landscape of biomedical research and gene therapy. Ongoing research in gene editing has led to the rapid iteration of CRISPR technologies, such as base and prime editors, enabling precise nucleotide changes without the need for generating harmful double-strand breaks (DSBs). Furthermore, innovations such as CRISPR fusion systems with DNA recombinases, DNA polymerases, and DNA ligases have expanded the size limitations for edited sequences, opening new avenues for therapeutic development. Beyond the CRISPR system, mobile genetic elements (MGEs) and epigenetic editors are emerging as efficient alternatives for precise large insertions or stable gene manipulation in mammalian cells. These advances collectively set the stage for next-generation gene therapy development. This review highlights recent developments of genetic and epigenetic editing tools and explores preclinical innovations poised to advance the field.
Collapse
Affiliation(s)
- Dongqi Liu
- Department of Pediatrics, Department of Molecular and Medical Genetics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Di Cao
- Department of Pediatrics, Department of Molecular and Medical Genetics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Renzhi Han
- Department of Pediatrics, Department of Molecular and Medical Genetics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
86
|
Manzoor S, Nabi SU, Ayaz A, Rasool B, Sharma SK, Chesti MH, Parveen S, Verma MK, Diab MA, Rabbee MF. Unveiling viral threats to temperate pome fruits: characterization, transmission, and sustainable management strategies. Front Microbiol 2025; 16:1560720. [PMID: 40177484 PMCID: PMC11961995 DOI: 10.3389/fmicb.2025.1560720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Apple (Malus × domestica Borkh.), pear (Pyrus communis L.), and quince (Cydonia oblonga Mill.) are widely cultivated fruit crops in temperate regions due to their desirable flavors and health benefits. However, their production is severely affected by various biotic stresses, with viral diseases being particularly significant challenge. These viral infections are of great economic importance, not only reduce tree vigor and yield but also compromise fruit quality and marketability. To date, more than 26 viruses and viroids have been identified as pathogens of these fruit trees. Many of these viral diseases persist as latent infections, causing permanent infections in these fruit trees. This review provides an overview of the viral pathogens affecting apple, pear, and quince, including their characterization, transmission modes, and the challenges they present for management. Emphasis is placed on accurate diagnosis and effective control strategies to mitigate the impact of these diseases in apple orchards.
Collapse
Affiliation(s)
- Subaya Manzoor
- Plant Virology Laboratory, ICAR-Central Institute of Temperate Horticulture Srinagar, Jammu and Kashmir, India
| | - Sajad Un Nabi
- Plant Virology Laboratory, ICAR-Central Institute of Temperate Horticulture Srinagar, Jammu and Kashmir, India
| | - Aadil Ayaz
- Plant Virology Laboratory, ICAR-Central Institute of Temperate Horticulture Srinagar, Jammu and Kashmir, India
| | - Bushra Rasool
- Plant Virology Laboratory, ICAR-Central Institute of Temperate Horticulture Srinagar, Jammu and Kashmir, India
| | - Susheel K. Sharma
- Division of Plant Pathology, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - M. H. Chesti
- Division of Soil Science and Agricultural Chemistry, SKUAST-K, Wadura, India
| | - Shugufta Parveen
- Apple Research Station, Pahnu Shopian, SKUAST-K, Shalimar, India
| | - M. K. Verma
- Plant Virology Laboratory, ICAR-Central Institute of Temperate Horticulture Srinagar, Jammu and Kashmir, India
| | - M. A. Diab
- Department of Biotechnology, Yeungnam University, Gyeongbuk, Gyeongsan, Republic of Korea
| | - Muhammad Fazle Rabbee
- Department of Biotechnology, Yeungnam University, Gyeongbuk, Gyeongsan, Republic of Korea
| |
Collapse
|
87
|
Wei B, Ma E, Tang S, Cadang L, Collins V, Gorman S, Chen B, Huang R, Wang J, Ma M, Zhang K. Real-Time Monitoring of Higher-Order Structure of RNAs by Temperature-Course Size Exclusion Chromatography and Microfluidic Modulation Spectroscopy. Anal Chem 2025; 97:5632-5642. [PMID: 40014844 DOI: 10.1021/acs.analchem.4c06343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Recently, there has been emerging interest in the characterization of the higher order structure (HOS) of oligonucleotide therapeutics because of its potential impact on the function. However, many existing experimental and computational methods face challenges with respect to throughput, cost, and resolution for large ribonucleic acids (RNAs). In this study, we present the use of two orthogonal analytical methods, size-exclusion chromatography (SEC) and microfluidic modulation spectroscopy (MMS), which are used to investigate conformational changes of two 100 mer single guide RNAs (sgRNAs) with complex HOS and aggregation specie profiles. SEC, coupled with multiangle light scattering (MALS), mass spectrometry (MS), and isothermal MMS revealed various forms of aggregation and potential interactions. We also developed temperature-course SEC and thermal ramping MMS methods to monitor real-time HOS changes from room temperature to the RNA melting point. Through the experiments, we observed two discrete steps of thermally induced dissociation of RNA aggregates, namely higher order aggregates (HOA) dissociation and dimer dissociation. Temperature-course SEC allows for thermodynamic analysis of the enthalpy and entropy of the reaction. We also identified two spectral regions in infrared (IR) spectra with thermal ramping MMS, 1665 cm-1 and between 1700 and 1720 cm-1, which closely correlated to the Watson-Crick base pairing and the related HOS change in RNA. The combination of SEC and MMS offers a comprehensive biophysical characterization toolkit for RNA HOS under native conditions, providing valuable insights for candidate optimization and formulation screening in the development of RNA therapeutics.
Collapse
Affiliation(s)
- Bingchuan Wei
- Synthetic Molecule Pharmaceutical Science, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Eugene Ma
- RedShift BioAnalytics, Inc., 80 Central Street, Boxborough, Massachusetts 01719, United States
| | - Shijia Tang
- Synthetic Molecule Pharmaceutical Science, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Lance Cadang
- Synthetic Molecule Pharmaceutical Science, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Valerie Collins
- RedShift BioAnalytics, Inc., 80 Central Street, Boxborough, Massachusetts 01719, United States
| | - Scott Gorman
- RedShift BioAnalytics, Inc., 80 Central Street, Boxborough, Massachusetts 01719, United States
| | - Bifan Chen
- Synthetic Molecule Pharmaceutical Science, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Richard Huang
- RedShift BioAnalytics, Inc., 80 Central Street, Boxborough, Massachusetts 01719, United States
| | - Jenny Wang
- Synthetic Molecule Pharmaceutical Science, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Maria Ma
- RedShift BioAnalytics, Inc., 80 Central Street, Boxborough, Massachusetts 01719, United States
| | - Kelly Zhang
- Synthetic Molecule Pharmaceutical Science, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
88
|
Ramadoss GN, Namaganda SJ, Hamilton JR, Sharma R, Chow KG, Macklin BL, Sun M, Liu JC, Fellmann C, Watry HL, Jin J, Perez BS, Espinoza CRS, Matia MP, Lu SH, Judge LM, Nussenzweig A, Adamson B, Murthy N, Doudna JA, Kampmann M, Conklin BR. Neuronal DNA repair reveals strategies to influence CRISPR editing outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.25.600517. [PMID: 38979269 PMCID: PMC11230251 DOI: 10.1101/2024.06.25.600517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Genome editing is poised to revolutionize treatment of genetic diseases, but poor understanding and control of DNA repair outcomes hinders its therapeutic potential. DNA repair is especially understudied in nondividing cells like neurons, which must withstand decades of DNA damage without replicating. This lack of knowledge limits the efficiency and precision of genome editing in clinically relevant cells. To address this, we used induced pluripotent stem cells (iPSCs) and iPSC-derived neurons to examine how postmitotic human neurons repair Cas9-induced DNA damage. We discovered that neurons can take weeks to fully resolve this damage, compared to just days in isogenic iPSCs. Furthermore, Cas9-treated neurons upregulated unexpected DNA repair genes, including factors canonically associated with replication. Manipulating this response with chemical or genetic perturbations allowed us to direct neuronal repair toward desired editing outcomes. By studying DNA repair in postmitotic human cells, we uncovered unforeseen challenges and opportunities for precise therapeutic editing.
Collapse
Affiliation(s)
- Gokul N Ramadoss
- Gladstone Institutes, San Francisco, CA, 94158, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, 94158, USA
| | | | - Jennifer R Hamilton
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Molecular & Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Rohit Sharma
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | | | | | - Mengyuan Sun
- Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Jia-Cheng Liu
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Christof Fellmann
- Gladstone Institutes, San Francisco, CA, 94158, USA
- Department of Cellular & Molecular Pharmacology, University of California, San Francisco, CA, 94158, USA
| | | | - Julianne Jin
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, 94158, USA
| | - Barbara S Perez
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Molecular & Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Cindy R Sandoval Espinoza
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Molecular & Cell Biology, University of California, Berkeley, CA, 94720, USA
| | | | - Serena H Lu
- Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Luke M Judge
- Gladstone Institutes, San Francisco, CA, 94158, USA
- Department of Pediatrics, University of California, San Francisco, CA, 94158, USA
| | - Andre Nussenzweig
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Britt Adamson
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
| | - Niren Murthy
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
| | - Jennifer A Doudna
- Gladstone Institutes, San Francisco, CA, 94158, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Molecular & Cell Biology, University of California, Berkeley, CA, 94720, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA, 94720, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA, 94720, USA
- Department of Chemistry, University of California, Berkeley, CA, 94720, USA
- MBIB Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, 94158, USA
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, 94158, USA
| | - Bruce R Conklin
- Gladstone Institutes, San Francisco, CA, 94158, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Department of Cellular & Molecular Pharmacology, University of California, San Francisco, CA, 94158, USA
- Department of Medicine, University of California, San Francisco, CA, 94158, USA
| |
Collapse
|
89
|
Peng W, Shi M, Hu B, Jia J, Li X, Wang N, Man S, Ye S, Ma L. Nanotechnology-leveraged CRISPR/Cas systems: icebreaking in trace cancer-related nucleic acids biosensing. Mol Cancer 2025; 24:78. [PMID: 40087758 PMCID: PMC11908094 DOI: 10.1186/s12943-024-02222-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/31/2024] [Indexed: 03/17/2025] Open
Abstract
As promising noninvasive biomarkers, nucleic acids provide great potential to innovate cancer early detection methods and promote subsequent diagnosis to improve the survival rates of patient. Accurate, straightforward and sensitive detection of such nucleic acid-based cancer biomarkers in complex biological samples holds significant clinical importance. However, the low abundance creates huge challenges for their routine detection. As the next-generation diagnostic tool, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein (Cas) with their high programmability, sensitivity, fidelity, single-base resolution, and precise nucleic acid positioning capabilities are extremely attractive for trace nucleic acid-based cancer biomarkers (NABCBs), permitting rapid, ultra-sensitive and specific detection. More importantly, by combing with nanotechnology, it can solve the long-lasting problems of poor sensitivity, accuracy and simplicity, as well as to achieve integrated miniaturization and portable point-of-care testing (POCT) detection. However, existing literature lacks specific emphasis on this topic. Thus, we intend to propose a timely one for the readers. This review will bridge this gap by providing insights for CRISPR/Cas-based nano-biosensing development and highlighting the current state-of-art, challenges, and prospects. We expect that it can provide better understanding and valuable insights for trace NABCBs detection, thereby facilitating advancements in early cancer screening/detection/diagnostics and win practical applications in the foreseeable future.
Collapse
Affiliation(s)
- Weipan Peng
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Mengting Shi
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Bin Hu
- Department of Pharmacy, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, China
| | - Jingyu Jia
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Xinyue Li
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Nan Wang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Shuli Man
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Shengying Ye
- Pharmacy Department, The 983th Hospital of The Joint Logistics Support Force of The Chinese People's Liberation Army, Tianjin, China.
| | - Long Ma
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Microbiology, Tianjin Key Laboratory of Industry Microbiology, International China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Ministry of Education, National and Local United Engineering Lab of Metabolic Control Fermentation Technology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| |
Collapse
|
90
|
Segretin ME, Soto GC, Lorenzo CD. Latin America: a hub for agrobiotechnological innovations. ANNALS OF BOTANY 2025; 135:629-642. [PMID: 39470392 PMCID: PMC11904903 DOI: 10.1093/aob/mcae191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Modern biotechnology is one of the last century's major advances in human science. Particularly in the agronomical field, the landscape of crop improvement technologies has witnessed a great expansion, driven by the integration of molecular and genetic engineering methodologies into the breeding toolbox. Latin America (LATAM) serves as a pioneering region in incorporating such techniques with several countries swiftly embracing these technologies. SCOPE This review aims to give a comprehensive overview of the elements that influenced agrobiotech acceptance in LATAM countries and how such cases could provide support for upcoming technologies to be considered worldwide. CONCLUSIONS Nearly 50 years of biotech breakthroughs have provided humankind with an impressive portfolio of tools already integrated into several life-science areas. The agronomical field has greatly progressed thanks to technologies derived from genetically modified organisms and great promises are being made to also incorporate genome-editing products. LATAM provides a prime example of how early introduction of novelties in the crop production chain can result in improved yields, paving the way for future developments to be easily integrated into the technological ecosystem of a region. The example set by LATAM can also be useful for the present gene-editing regulatory scenario. With several countries presently on the path to approving these methods in their current crop systems, basing their next steps on the example of LATAM could represent a safe and practical pathway towards a new agronomical revolution.
Collapse
Affiliation(s)
- Maria Eugenia Segretin
- Laboratorio de Biotecnología Vegetal, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular-INGEBI-CONICET, Vuelta de Obligado 2490, C1428ADN, Ciudad Autónoma de Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Intendente Güiraldes 2160, Ciudad Autónoma Buenos Aires, C1428EGA, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 (C1425FQB) Ciudad Autónoma de Buenos Aires, Argentina
| | - Gabriela Cynthia Soto
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 (C1425FQB) Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Genética ‘Ewald Favret’ (INTA), Buenos Aires, Argentina
| | - Christian Damian Lorenzo
- Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Gent, Belgium
- Center for Plant Systems Biology, VIB, B-9052 Gent, Belgium
| |
Collapse
|
91
|
Park J, Rhoo KY, Kim Y, Kim YS, Paik SR. Cell-Division-Independent Rapid Expression of DNA Delivered with α-Synuclein-Gold Nanoparticle Conjugates. ACS APPLIED MATERIALS & INTERFACES 2025; 17:14846-14858. [PMID: 40014054 DOI: 10.1021/acsami.4c17967] [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: 02/28/2025]
Abstract
Gene delivery is a primary technology employed in diverse areas of biomedical science, from gene therapy to gene editing, cancer treatment, and stem cell research. Here, we introduce a gene delivery system utilizing an intrinsically disordered protein of α-synuclein (αS) demonstrated to interact with lipid membranes by transforming its original random structure to an α-helix. Since the helix bundle formation is a signature of cell-penetrating peptides for membrane translocation, a multitude of αS(Y136C)s replacing tyrosine at the C-terminus with cysteine were covalently attached onto gold nanoparticles (AuNPs) in a specific orientation with the helix-forming basic N-termini exposed outward. The resulting αS(Y136C)-AuNP conjugates were found to exhibit a rapid gene expression without causing cytotoxicity when the gene of the enhanced green fluorescent protein (EGFP) was delivered with the conjugates into the cells. Based on inhibition studies toward endocytosis and mitosis, the αS(Y136C)-AuNP/DNA complex was demonstrated to take both endosomal and non-endosomal intracellular transport pathways. The DNA translocation into the nucleus was independent of cell division. This nondisruptive and rapid DNA transfection by αS(Y136C)-AuNPs allowed a successful delivery of granzyme A gene leading to cellular pyroptosis. Modifications of αS(Y136C)-AuNP/DNA complex, such as antibody immobilization and replacement of DNA with biological suprastructures including RNA, protein, and nonbiological fusion materials, would allow the intracellular delivery system to be applied in diverse areas of future biotechnology.
Collapse
Affiliation(s)
- Jeongha Park
- School of Chemical and Biological Engineering, Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Kun Yil Rhoo
- Interdisciplinary Program of Bioengineering, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Yunsoo Kim
- School of Chemical and Biological Engineering, Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Sik Kim
- Department of Pathology, Ansan Hospital, Korea University College of Medicine, Ansan 15355, Republic of Korea
| | - Seung R Paik
- School of Chemical and Biological Engineering, Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program of Bioengineering, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
92
|
Hunt JM, du Rand A, Verdon D, Clemance L, Loef E, Malhi C, Buttle B, Knapp DJ, Michaels YS, Garlick J, Dunbar PR, Purvis D, Feisst V, Sheppard H. Enhanced HDR-mediated correction of heterozygous COL7A1 mutations for recessive dystrophic epidermolysis bullosa. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102472. [PMID: 40027886 PMCID: PMC11872078 DOI: 10.1016/j.omtn.2025.102472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
Gene editing facilitated by homology-directed repair (HDR) holds great potential for treating monogenetic disorders such as recessive dystrophic epidermolysis bullosa (RDEB). However, low efficiency and variability between loci must be overcome for its widespread adoption into personalized therapies. To address these challenges, we developed a highly efficient and versatile gene editing strategy for RDEB that incorporates the small molecule inhibitor M3814 to enhance HDR. We focused on three RDEB causative COL7A1 mutations not previously targeted by existing gene therapies. Editing was achieved using Cas9-nuclease ribonucleoproteins with short single-stranded DNA donor templates, and outcomes were assessed with an Oxford Nanopore Technology sequencing analysis pipeline. We demonstrate precise genomic HDR rates of up to 75% of alleles in primary RDEB keratinocytes and 32% in fibroblasts. This approach restored collagen VII expression in up to 80% of keratinocytes within a bulk-edited population and resulted in correct collagen VII deposition in a 3D skin model. Additionally, at one locus we show that a dual Cas9-nickase strategy is less effective than Cas9-nuclease and prone to large on-target deletions. Our results demonstrate a significant advancement in the efficiency and consistency of HDR editing, potentially paving the way for more effective personalized gene therapies.
Collapse
Affiliation(s)
- John M.T. Hunt
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
- Maurice Wilkins Center, New Zealand
| | - Alex du Rand
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Daniel Verdon
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Leah Clemance
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Evert Loef
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Chloe Malhi
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Ben Buttle
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - David J.H.F. Knapp
- Institut de recherche en immunologie et en cancérologie (IRIC) and Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Yale S. Michaels
- Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB R2H 2A6, Canada
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Jonathan Garlick
- School of Dental Medicine, Tufts University, Boston, MA 02111, USA
| | - P. Rod Dunbar
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
- Maurice Wilkins Center, New Zealand
| | - Diana Purvis
- Te Whatu Ora Health New Zealand, Te Toka Tumai, Auckland 1023, New Zealand
| | - Vaughan Feisst
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
| | - Hilary Sheppard
- School of Biological Sciences, The University of Auckland, Auckland 1010, New Zealand
- Maurice Wilkins Center, New Zealand
| |
Collapse
|
93
|
Ouaarous M, El Fakhouri K, Taarji N, Baouchi A, Amri M, Ramdani C, Sobeh M, Mesfioui A, El Bouhssini M. Impact of Field Insect Pests on Seed and Nutritional Quality of Some Important Crops: A Comprehensive Review. ACS OMEGA 2025; 10:8779-8792. [PMID: 40092757 PMCID: PMC11904676 DOI: 10.1021/acsomega.4c08982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/26/2024] [Accepted: 01/17/2025] [Indexed: 03/19/2025]
Abstract
The increasing problem of insect pest infestation in agriculture and its impact on crop production and productivity require a thorough understanding and identification of efficient control solutions. This review explores the effects of insect infestations on crop productivity and seed quality with a focus on nutritional value, in particular, protein content, sugar levels, mineral composition, vitamin C concentration, antioxidant activity, and phenolic content. The paper compiles current studies on the diverse impacts of insect pests, including Hessian flies, Russian wheat aphids, Sunn pests, fruit flies, and others, on various crops such as wheat, soybeans, mung beans, durum wheat, bread wheat, olives, and citrus fruits. Integrated pest management (IPM) techniques including the implementation of digital technologies are highlighted as crucial methods for maintaining crop quality and ensuring food and nutritional security. This review provides more emphasis on seed morphological and nutritional quality characterization in response to insect pest infestation as well as the development of more practical methods for lowering crop quality losses and raising the nutritional quality of agricultural products by adopting a multidimensional approach for durable insect pest management.
Collapse
Affiliation(s)
- Mohamed Ouaarous
- AgroBioSciences
Program, College of Agriculture and Environmental Sciences, Mohammed VI Polytechnic University, Lot 660, Hay Moulay Rachid, 43150 Benguerir, Morocco
- Laboratory
of Biology and Health, Department of Biology, Faculty of Science, Ibn-Tofail University, Kenitra 14000, Morocco
| | - Karim El Fakhouri
- AgroBioSciences
Program, College of Agriculture and Environmental Sciences, Mohammed VI Polytechnic University, Lot 660, Hay Moulay Rachid, 43150 Benguerir, Morocco
| | - Noamane Taarji
- AgroBioSciences
Program, College of Agriculture and Environmental Sciences, Mohammed VI Polytechnic University, Lot 660, Hay Moulay Rachid, 43150 Benguerir, Morocco
| | - Adil Baouchi
- AgroBioSciences
Program, College of Agriculture and Environmental Sciences, Mohammed VI Polytechnic University, Lot 660, Hay Moulay Rachid, 43150 Benguerir, Morocco
| | - Moez Amri
- AgroBioSciences
Program, College of Agriculture and Environmental Sciences, Mohammed VI Polytechnic University, Lot 660, Hay Moulay Rachid, 43150 Benguerir, Morocco
| | - Chaimae Ramdani
- AgroBioSciences
Program, College of Agriculture and Environmental Sciences, Mohammed VI Polytechnic University, Lot 660, Hay Moulay Rachid, 43150 Benguerir, Morocco
| | - Mansour Sobeh
- AgroBioSciences
Program, College of Agriculture and Environmental Sciences, Mohammed VI Polytechnic University, Lot 660, Hay Moulay Rachid, 43150 Benguerir, Morocco
| | - Abdelhalem Mesfioui
- Laboratory
of Biology and Health, Department of Biology, Faculty of Science, Ibn-Tofail University, Kenitra 14000, Morocco
| | - Mustapha El Bouhssini
- AgroBioSciences
Program, College of Agriculture and Environmental Sciences, Mohammed VI Polytechnic University, Lot 660, Hay Moulay Rachid, 43150 Benguerir, Morocco
| |
Collapse
|
94
|
Chavhan RL, Jaybhaye SG, Hinge VR, Deshmukh AS, Shaikh US, Jadhav PK, Kadam US, Hong JC. Emerging applications of gene editing technologies for the development of climate-resilient crops. Front Genome Ed 2025; 7:1524767. [PMID: 40129518 PMCID: PMC11931038 DOI: 10.3389/fgeed.2025.1524767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/07/2025] [Indexed: 03/26/2025] Open
Abstract
Climate change threatens global crop yield and food security due to rising temperatures, erratic rainfall, and increased abiotic stresses like drought, heat, and salinity. Gene editing technologies, including CRISPR/Cas9, base editors, and prime editors, offer precise tools for enhancing crop resilience. This review explores the mechanisms of these technologies and their applications in developing climate-resilient crops to address future challenges. While CRISPR/enables targeted modifications of plant DNA, the base editors allow for direct base conversion without inducing double-stranded breaks, and the prime editors enable precise insertions, deletions, and substitutions. By understanding and manipulating key regulator genes involved in stress responses, such as DREB, HSP, SOS, ERECTA, HsfA1, and NHX; crop tolerance can be enhanced against drought, heat, and salt stress. Gene editing can improve traits related to root development, water use efficiency, stress response pathways, heat shock response, photosynthesis, membrane stability, ion homeostasis, osmotic adjustment, and oxidative stress response. Advancements in gene editing technologies, integration with genomics, phenomics, artificial intelligence (AI)/machine learning (ML) hold great promise. However, challenges such as off-target effects, delivery methods, and regulatory barriers must be addressed. This review highlights the potential of gene editing to develop climate-resilient crops, contributing to food security and sustainable agriculture.
Collapse
Affiliation(s)
- R. L. Chavhan
- Vilasrao Deshmukh College of Agricultural Biotechnology, Vasantrao Naik Marathwada Krishi Vidyapeeth, Latur, India
| | - S. G. Jaybhaye
- Vilasrao Deshmukh College of Agricultural Biotechnology, Vasantrao Naik Marathwada Krishi Vidyapeeth, Latur, India
| | - V. R. Hinge
- Vilasrao Deshmukh College of Agricultural Biotechnology, Vasantrao Naik Marathwada Krishi Vidyapeeth, Latur, India
| | - A. S. Deshmukh
- Vilasrao Deshmukh College of Agricultural Biotechnology, Vasantrao Naik Marathwada Krishi Vidyapeeth, Latur, India
| | - U. S. Shaikh
- Vilasrao Deshmukh College of Agricultural Biotechnology, Vasantrao Naik Marathwada Krishi Vidyapeeth, Latur, India
| | - P. K. Jadhav
- Vilasrao Deshmukh College of Agricultural Biotechnology, Vasantrao Naik Marathwada Krishi Vidyapeeth, Latur, India
| | - U. S. Kadam
- Division of Applied Life Science (BK21 Four), Division of Life Science, Plant Molecular Biology and Biotechnology Research Centre (PMBBRC), Gyeongsang National University, Jinju, Republic of Korea
| | - J. C. Hong
- Division of Applied Life Science (BK21 Four), Division of Life Science, Plant Molecular Biology and Biotechnology Research Centre (PMBBRC), Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
95
|
Chen X, Wang Y, Zhang Y, Li X, Zhang L, Gao S, Zhang C. Neural Excitatory/Inhibitory Imbalance in Motor Aging: From Genetic Mechanisms to Therapeutic Challenges. BIOLOGY 2025; 14:272. [PMID: 40136528 PMCID: PMC11939721 DOI: 10.3390/biology14030272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025]
Abstract
Neural excitatory/inhibitory (E/I) imbalance plays a pivotal role in the aging process. However, despite its significant impact, the role of E/I imbalance in motor dysfunction and neurodegenerative diseases has not received sufficient attention. This review explores the mechanisms underlying motor aging through the lens of E/I balance, emphasizing genetic and molecular factors that contribute to this imbalance (such as SCN2A, CACNA1C, GABRB3, GRIN2A, SYT, BDNF…). Key regulatory genes, including REST, vps-34, and STXBP1, are examined for their roles in modulating synaptic activity and neuronal function during aging. With insights drawn from ALS, we discuss how disruptions in E/I balance contribute to the pathophysiology of age-related motor dysfunction. The genes discussed above exhibit a certain association with age-related motor neuron diseases (like ALS), a relationship that had not been previously recognized. Innovative genetic therapies, such as gene editing technology and optogenetic manipulation, are emerging as promising tools for restoring E/I balance, offering hope for ameliorating motor deficits in aging. This review explores the potential of these technologies to intervene in aging-related motor diseases, despite challenges in their direct application to human conditions.
Collapse
Affiliation(s)
- Xuhui Chen
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.C.); (L.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ya Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.W.); (Y.Z.); (X.L.)
| | - Yongning Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.W.); (Y.Z.); (X.L.)
| | - Xucheng Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.W.); (Y.Z.); (X.L.)
| | - Le Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.C.); (L.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shangbang Gao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.C.); (L.Z.)
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.W.); (Y.Z.); (X.L.)
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.C.); (L.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
96
|
Casco-Robles MM, Echigoya T, Shimazaki T, Murakami Y, Hirano M, Maruo F, Mizuno S, Takahashi S, Chiba C. One-step Cre-loxP organism creation by TAx9. Commun Biol 2025; 8:340. [PMID: 40050380 PMCID: PMC11885649 DOI: 10.1038/s42003-025-07759-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/17/2025] [Indexed: 03/09/2025] Open
Abstract
The creation of organisms with Cre-loxP conditional gene recombination systems often faces challenges, particularly when creating the initial (F0) generation with both a Cre recombinase and a DNA site flanked by loxP elements (floxed site). The primary reason is that it is difficult to synthesize a single plasmid with both the Cre gene and the floxed site, since Cre-mediated recombination spontaneously occurs when the plasmid is amplified in Escherichia coli bacterial cells. Here, we introduce an artificial nucleic acid sequence TATATATATATATATATA, named TAx9, that enables the integration of both the Cre gene and the floxed site into a single plasmid. TAx9 effectively blocks spontaneous Cre-mediated recombination in E. coli cells. Using this system, we created an F0 generation of transgenic newts and CRISPR-Cas9 knock-in mice with tissue-specific Cre recombination triggered by tamoxifen. TAx9 technology will be a powerful strategy for creating organisms capable of conditional genetic modification in the F0 generation, accelerating various life science research by reducing the time and cost for ultimately establishing and maintaining lines of genetically modified organisms.
Collapse
Affiliation(s)
| | - Tomoki Echigoya
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takeaki Shimazaki
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuri Murakami
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Masaya Hirano
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Fumiaki Maruo
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Anatomy and Embryology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Chikafumi Chiba
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
97
|
García-Tenorio EM, Álvarez M, Gallego-Bonhomme M, Desviat LR, Richard E. Novel CRISPR-Cas9 iPSC knockouts for PCCA and PCCB genes: advancing propionic acidemia research. Hum Cell 2025; 38:64. [PMID: 40044943 PMCID: PMC11882705 DOI: 10.1007/s13577-025-01193-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 02/25/2025] [Indexed: 03/09/2025]
Abstract
Propionic acidemia (PA) is a rare autosomal recessive metabolic disorder caused by mutations in the PCCA and PCCB genes, which encode subunits of the mitochondrial enzyme propionyl-CoA carboxylase (PCC). This enzyme deficiency leads to the accumulation of toxic metabolites, resulting in severe metabolic dysfunction. To create ideal in vitro disease models of PA with isogenic controls and provide a robust platform for therapeutic research, we generated two induced pluripotent stem cell (iPSC) lines with knockout (KO) mutations in the PCCA and PCCB genes using CRISPR-Cas9 gene editing in a healthy control iPSC line. The KO iPS cells were successfully established and characterized, confirming the presence of frameshift insertions and deletions in each target gene, as well as the loss of the corresponding transcript, protein expression, and activity. Additionally, the generated iPSC lines exhibit hallmark characteristics of pluripotency, including the potential to differentiate into all three germ layers. Our PCCA and PCCB KO iPSC models provide a valuable tool for studying the molecular mechanisms underlying PA and hold potential for advancing new therapeutic approaches.
Collapse
Affiliation(s)
- Emilio M García-Tenorio
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Instituto Universitario de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Mar Álvarez
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | | | - Lourdes R Desviat
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Instituto Universitario de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), Madrid, Spain
| | - Eva Richard
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049, Madrid, Spain.
- Instituto Universitario de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain.
- Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), Madrid, Spain.
| |
Collapse
|
98
|
Li M, Wu L, Si H, Wu Y, Liu Y, Zeng Y, Shen B. Engineered mitochondria in diseases: mechanisms, strategies, and applications. Signal Transduct Target Ther 2025; 10:71. [PMID: 40025039 PMCID: PMC11873319 DOI: 10.1038/s41392-024-02081-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/30/2024] [Accepted: 11/17/2024] [Indexed: 03/04/2025] Open
Abstract
Mitochondrial diseases represent one of the most prevalent and debilitating categories of hereditary disorders, characterized by significant genetic, biological, and clinical heterogeneity, which has driven the development of the field of engineered mitochondria. With the growing recognition of the pathogenic role of damaged mitochondria in aging, oxidative disorders, inflammatory diseases, and cancer, the application of engineered mitochondria has expanded to those non-hereditary contexts (sometimes referred to as mitochondria-related diseases). Due to their unique non-eukaryotic origins and endosymbiotic relationship, mitochondria are considered highly suitable for gene editing and intercellular transplantation, and remarkable progress has been achieved in two promising therapeutic strategies-mitochondrial gene editing and artificial mitochondrial transfer (collectively referred to as engineered mitochondria in this review) over the past two decades. Here, we provide a comprehensive review of the mechanisms and recent advancements in the development of engineered mitochondria for therapeutic applications, alongside a concise summary of potential clinical implications and supporting evidence from preclinical and clinical studies. Additionally, an emerging and potentially feasible approach involves ex vivo mitochondrial editing, followed by selection and transplantation, which holds the potential to overcome limitations such as reduced in vivo operability and the introduction of allogeneic mitochondrial heterogeneity, thereby broadening the applicability of engineered mitochondria.
Collapse
Affiliation(s)
- Mingyang Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Limin Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Haibo Si
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuangang Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuan Liu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yi Zeng
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Bin Shen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
99
|
Barber HM, Pater AA, Gagnon KT, Damha MJ, O'Reilly D. Chemical engineering of CRISPR-Cas systems for therapeutic application. Nat Rev Drug Discov 2025; 24:209-230. [PMID: 39690326 DOI: 10.1038/s41573-024-01086-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 12/19/2024]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) technology has transformed molecular biology and the future of gene-targeted therapeutics. CRISPR systems comprise a CRISPR-associated (Cas) endonuclease and a guide RNA (gRNA) that can be programmed to guide sequence-specific binding, cleavage, or modification of complementary DNA or RNA. However, the application of CRISPR-based therapeutics is challenged by factors such as molecular size, prokaryotic or phage origins, and an essential gRNA cofactor requirement, which impact efficacy, delivery and safety. This Review focuses on chemical modification and engineering approaches for gRNAs to enhance or enable CRISPR-based therapeutics, emphasizing Cas9 and Cas12a as therapeutic paradigms. Issues that chemically modified gRNAs seek to address, including drug delivery, physiological stability, editing efficiency and off-target effects, as well as challenges that remain, are discussed.
Collapse
Affiliation(s)
- Halle M Barber
- Department of Chemistry, McGill University, Montreal, Quebec, Canada
| | - Adrian A Pater
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Keith T Gagnon
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | - Masad J Damha
- Department of Chemistry, McGill University, Montreal, Quebec, Canada.
| | - Daniel O'Reilly
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA.
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
100
|
Wang Y, Tian Y, Xu D, Cheng S, Li WW, Song H. Recent advances in synthetic biology toolkits and metabolic engineering of Ralstonia eutropha H16 for production of value-added chemicals. Biotechnol Adv 2025; 79:108516. [PMID: 39793936 DOI: 10.1016/j.biotechadv.2025.108516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Ralstonia eutropha H16, a facultative chemolithoautotrophic Gram-negative bacterium, demonstrates remarkable metabolic flexibility by utilizing either diverse organic substrates or CO2 as the sole carbon source, with H2 serving as the electron donor under aerobic conditions. The capacity of carbon and energy metabolism of R. eutropha H16 enabled development of synthetic biology technologies and strategies to engineer its metabolism for biosynthesis of value-added chemicals. This review firstly outlines the development of synthetic biology tools tailored for R. eutropha H16, including construction of expression vectors, regulatory elements, and transformation techniques. The availability of comprehensive omics data (i.e., transcriptomic, proteomic, and metabolomic) combined with the fully annotated genome sequence provides a robust genetic framework for advanced metabolic engineering. These advancements facilitate efficient reprogramming metabolic network of R. eutropha. The potential of R. eutropha as a versatile microbial platform for industrial biotechnology is further underscored by its ability to utilize a wide range of carbon sources for the production of value-added chemicals through both autotrophic and heterotrophic pathways. The integration of state-of-the-art genetic and genomic engineering tools and strategies with high cell-density fermentation processes enables engineered R. eutropha as promising microbial cell factories for optimizing carbon fluxes and expanding the portfolio of bio-based products.
Collapse
Affiliation(s)
- Ye Wang
- State Key Laboratory of Synthetic Biology, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Yao Tian
- State Key Laboratory of Synthetic Biology, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Dake Xu
- Shenyang National Laboratory for Materials Science, Northeastern University, 110819 Shenyang, China; Electrobiomaterials Institute, Key Laboratory for Anisotropy and Texture of Materials (Ministry of Education), Northeastern University, 110819 Shenyang, China
| | - Shaoan Cheng
- State Key Laboratory of Clean Energy, Department of Energy Engineering, Zhejiang University, Hangzhou 310027, China
| | - Wen-Wei Li
- Chinese Academy of Sciences Key Laboratory of Urban Pollutant Conversion, Department of Environmental Science and Engineering, University of Science & Technology of China, Hefei 230026, China
| | - Hao Song
- State Key Laboratory of Synthetic Biology, and School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; College of Life and Health Sciences, Northeastern University, Shenyang 110169, China.
| |
Collapse
|