101
|
Hernandez FJ. Nucleases: From Primitive Immune Defenders to Modern Biotechnology Tools. Immunology 2025; 174:279-286. [PMID: 39686519 PMCID: PMC11799398 DOI: 10.1111/imm.13884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/22/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
The story of nucleases begins on the ancient battlefields of early Earth, where simple bacteria fought to survive against viral invaders. Nucleases are enzymes that degrade nucleic acids, with restriction endonucleases emerging as some of the earliest defenders, cutting foreign DNA to protect their bacteria hosts. However, bacteria sought more than just defence. They evolved the CRISPR-Cas system, an adaptive immune mechanism capable of remembering past invaders. The now-famous Cas9 nuclease, a key player in this system, has been harnessed for genome editing, revolutionising biotechnology. Over time, nucleases evolved from basic viral defence tools into complex regulators of immune function in higher organisms. In humans, DNases and RNases maintain immune balance by clearing cellular debris, preventing autoimmunity, and defending against pathogens. These enzymes have transformed from simple bacterial defenders to critical players in both human immunity and biotechnology. This review explores the evolutionary history of nucleases and their vital roles as protectors in the story of life's defence mechanisms.
Collapse
Affiliation(s)
- Frank J. Hernandez
- Department of Physics, Chemistry and BiologyLinköping UniversityLinköpingSweden
- Department of Bioengineering and Biosciences, TECNUNNavarra UniversityDonostiaSpain
- IKERBASQUE, Basque Foundation for ScienceBilbaoSpain
| |
Collapse
|
102
|
Perez AR, Mavrothalassitis O, Chen JS, Hellman J, Gropper MA. CRISPR: fundamental principles and implications for anaesthesia. Br J Anaesth 2025; 134:839-852. [PMID: 39855935 PMCID: PMC11867086 DOI: 10.1016/j.bja.2024.11.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/22/2024] [Accepted: 11/01/2024] [Indexed: 01/27/2025] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-based medical therapies are increasingly gaining regulatory approval worldwide. Consequently, patients receiving CRISPR therapy will come under the care of anaesthesiologists. An understanding of CRISPR, its technological implementations, and the characteristics of patients likely to receive this therapy will be essential to caring for this patient population. However, the role of CRISPR in anaesthesiology extends beyond simply caring for patients with prior CRISPR therapy. CRISPR has multiple direct potential applications in anaesthesia, particularly for managing chronic pain and critical illness. Additionally, given the unique skills anaesthesiologists possess, CRISPR potentially allows new roles for anaesthesiologists in the field of oncology. Consequently, CRISPR technology could enable new domains of anaesthetic practice. This review provides a primer on CRISPR for anaesthesiologists and an overview on how the technology could impact the field.
Collapse
Affiliation(s)
- Alexendar R Perez
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Silico Therapeutics, Inc., San Jose, CA, USA.
| | - Orestes Mavrothalassitis
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | | | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Michael A Gropper
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
103
|
Schwartz MK, Dunn SL, Gendron WAC, Helm JE, Kamau WS, Mark-Shadbolt M, Moehrenschlager A, Redford KH, Russell G, Sandler RL, Schultz CA, Wiedenheft B, Emmel AS, Brodie JF. Principles for introducing new genes and species for conservation. Trends Ecol Evol 2025; 40:296-307. [PMID: 39667987 DOI: 10.1016/j.tree.2024.11.011] [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: 04/28/2024] [Revised: 10/29/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024]
Abstract
Introducing new genes and new species into ecosystems where they have not previously existed presents opportunities and complex, multivalue decisions for conservation biologists and the public. Both synthetic biology and conservation introductions offer potential benefits, such as avoiding extinctions and restoring ecological function, but also carry risks of unintended ecological consequences and raise social and moral concerns. Although the conservation community has attempted to establish guidelines for each new tool, there is a need for comprehensive principles that will enable conservation managers to navigate emerging technologies. Here, we combine biological, legal, social, cultural, and ethical considerations into an inclusive set of principles designed to facilitate the efforts of managers facing high-consequence conservation decisions by clarifying the stakes of inaction and action, along with the use of decision frameworks to integrate multiple considerations.
Collapse
Affiliation(s)
- Michael K Schwartz
- USDA Forest Service, Rocky Mountain Research Station, Missoula, MT, USA.
| | - Summer L Dunn
- USDA Forest Service, Policy Office, Strategy and Analysis Branch, Washington, DC, USA
| | - William A C Gendron
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Jennifer E Helm
- USDA Forest Service, Rocky Mountain Research Station, Missoula, MT, USA
| | - W Sebastian Kamau
- Media Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | | | - Gregory Russell
- Department of Forest and Rangeland Stewardship, Colorado State University, Fort Collins, CO, USA
| | - Ronald L Sandler
- The Ethics Institute and Department of Philosophy and Religion, Northeastern University, Boston, MA, USA
| | - Courtney A Schultz
- Department of Forest and Rangeland Stewardship, Colorado State University, Fort Collins, CO, USA
| | - Blake Wiedenheft
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Amanda S Emmel
- Wildlife Biology Program, University of Montana, Missoula, MT, USA
| | - Jedediah F Brodie
- Wildlife Biology Program, University of Montana, Missoula, MT, USA; Division of Biological Sciences and Wildlife Biology Program, University of Montana, Missoula, MT, USA; Institute for Biodiversity and Environmental Conservation, Universiti Malaysia Sarawak, Kota Samarahan, Sarawak, Malaysia
| |
Collapse
|
104
|
Chaugule S, Constantinou CK, John AA, Micha D, Eekhoff M, Gravallese E, Gao G, Shim JH. Comprehensive Review of Osteogenesis Imperfecta: Current Treatments and Future Innovations. Hum Gene Ther 2025; 36:597-617. [PMID: 39932815 PMCID: PMC11971546 DOI: 10.1089/hum.2024.191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/08/2025] [Indexed: 02/13/2025] Open
Abstract
Osteogenesis imperfecta (OI) is a rare genetic disorder characterized by bone fragility due to reduced bone quality, often accompanied by low bone mass, recurrent fractures, hearing loss, skeletal abnormalities, and short stature. Pathogenic variants in over 20 genes lead to clinical and genetic variability in OI, resulting in diverse symptoms and severity. Current management involves a multidisciplinary approach, including antiresorptive medications, physiotherapy, occupational therapy, and orthopedic surgery, which provide symptomatic relief but no cure. Advancements in gene therapy technologies and stem cell therapies offer promising prospects for long-lasting or permanent solutions. This review provides a comprehensive overview of OI's classification, pathogenesis, and current treatment options. It also explores emerging biotechnologies for stem cells and gene-targeted therapies in OI. The potential of these innovative therapies and their clinical implementation challenges are evaluated, focusing on their imminent success in treating bone disorders.
Collapse
Affiliation(s)
- Sachin Chaugule
- Division of Rheumatology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | | | - Aijaz Ahmad John
- Division of Rheumatology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Dimitra Micha
- Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam; Amsterdam Rare Bone Disease center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam Reproduction and Development, Amsterdam, The Netherlands
| | - Marelise Eekhoff
- Department of Internal Medicine, Section Endocrinology & Metabolism, Amsterdam UMC, Vrije Universiteit Amsterdam; Amsterdam Rare Bone Disease center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam Reproduction and Development Amsterdam, Amsterdam, Netherlands
| | - Ellen Gravallese
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Jae-Hyuck Shim
- Division of Rheumatology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Genetic and Cellular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
105
|
Peters Haugrud AR, Achilli AL, Martínez‐Peña R, Klymiuk V. Future of durum wheat research and breeding: Insights from early career researchers. THE PLANT GENOME 2025; 18:e20453. [PMID: 38760906 PMCID: PMC11733671 DOI: 10.1002/tpg2.20453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/26/2024] [Accepted: 04/02/2024] [Indexed: 05/20/2024]
Abstract
Durum wheat (Triticum turgidum ssp. durum) is globally cultivated for pasta, couscous, and bulgur production. With the changing climate and growing world population, the need to significantly increase durum production to meet the anticipated demand is paramount. This review summarizes recent advancements in durum research, encompassing the exploitation of existing and novel genetic diversity, exploration of potential new diversity sources, breeding for climate-resilient varieties, enhancements in production and management practices, and the utilization of modern technologies in breeding and cultivar development. In comparison to bread wheat (T. aestivum), the durum wheat community and production area are considerably smaller, often comprising many small-family farmers, notably in African and Asian countries. Public breeding programs such as the International Maize and Wheat Improvement Center (CIMMYT) and the International Center for Agricultural Research in the Dry Areas (ICARDA) play a pivotal role in providing new and adapted cultivars for these small-scale growers. We spotlight the contributions of these and others in this review. Additionally, we offer our recommendations on key areas for the durum research community to explore in addressing the challenges posed by climate change while striving to enhance durum production and sustainability. As part of the Wheat Initiative, the Expert Working Group on Durum Wheat Genomics and Breeding recognizes the significance of collaborative efforts in advancing toward a shared objective. We hope the insights presented in this review stimulate future research and deliberations on the trajectory for durum wheat genomics and breeding.
Collapse
Affiliation(s)
- Amanda R. Peters Haugrud
- Cereal Crops Research Unit, Edward T. Schafer Agricultural Research Center, Agricultural Research ServiceUnited States Department of AgricultureFargoNorth DakotaUSA
| | - Ana Laura Achilli
- Faculty of Land and Food SystemsThe University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Raquel Martínez‐Peña
- Regional Institute of Agri‐Food and Forestry Research and Development of Castilla‐La Mancha (IRIAF)Agroenvironmental Research Center El ChaparrilloCiudad RealSpain
| | - Valentyna Klymiuk
- Crop Development Centre and Department of Plant SciencesUniversity of SaskatchewanSaskatoonSaskatchewanCanada
| |
Collapse
|
106
|
Pavan-Guimaraes J, Devos L, Lascaris B, de Meijer VE, Monbaliu D, Jochmans I, Pulitano C, Porte RJ, Martins PN. Long-Term Liver Machine Perfusion Preservation: A Review of Recent Advances, Benefits and Logistics. Artif Organs 2025; 49:339-352. [PMID: 39895504 DOI: 10.1111/aor.14941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/23/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND The global shortage of suitable donor livers for transplantation has prompted efforts to expand the donor pool by using extended criteria donors. Machine preservation technology has shown promise in optimizing graft preservation and improving logistics. Additionally, it holds potential for organ repair, regeneration, therapeutic applications during extended preservation periods, and enhancing organ allocation. METHODS We conducted a comprehensive literature review using PubMed, Embase, and Web of Science databases. All studies published between January 1, 2022, and February 7, 2024, that described machine perfusion preservation of livers for more than 24 h were eligible for inclusion. The findings were synthesized in a narrative review format to highlight key benefits and advancements. RESULTS We identified eleven studies from multiple research groups, employing various techniques, devices, and preservation durations. Perfusion durations ranged from 1 to 13 days, with notable variations in protocols for long-term preservation beyond 24 h. Viability was assessed during perfusion only. No livers were transplanted. Among the reviewed studies, the introduction of a dialysis system emerged as the most effective strategy for managing waste accumulation during long-term liver perfusion. Differences were also observed in hemodynamics, oxygenation, organ chambers, supplemental regimens, and glycemic control. CONCLUSION Over the past two years, substantial progress has been made in refining protocols for long-term liver machine perfusion, with significant advancements in waste management, enabling successful multi-day perfusions. While these developments are promising, further research is necessary to standardize and optimize long-term perfusion protocols, establishing a reliable platform for both organ preservation and therapeutic applications.
Collapse
Affiliation(s)
| | - Lene Devos
- Department of Microbiology, Immunology and Transplantation, Transplantation Research Group, Lab of Abdominal Transplantation, KU Leuven, Leuven, Belgium
| | - Bianca Lascaris
- Section of HPB Surgery and Liver Transplantation, UMCG Comprehensive Transplant Center, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Vincent E de Meijer
- Section of HPB Surgery and Liver Transplantation, UMCG Comprehensive Transplant Center, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands
| | - Diethard Monbaliu
- Department of Microbiology, Immunology and Transplantation, Transplantation Research Group, Lab of Abdominal Transplantation, KU Leuven, Leuven, Belgium
- Abdominal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Ina Jochmans
- Department of Microbiology, Immunology and Transplantation, Transplantation Research Group, Lab of Abdominal Transplantation, KU Leuven, Leuven, Belgium
- Abdominal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Carlo Pulitano
- Australian National Liver Transplantation Unit, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Robert J Porte
- Division of HPB and Transplant Surgery, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Paulo N Martins
- Department of Surgery, Transplant Institute, University of Oklahoma, Oklahoma City, Oklahoma, USA
| |
Collapse
|
107
|
van Dijk A, Wilson AM, Marx B, Hough B, Swalarsk-Parry B, De Vos L, Wingfield MJ, Wingfield BD, Steenkamp ET. CRISPR-Cas9 genome editing reveals that the Pgs gene of Fusarium circinatum is involved in pathogenicity, growth and sporulation. Fungal Genet Biol 2025; 177:103970. [PMID: 39952463 DOI: 10.1016/j.fgb.2025.103970] [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: 06/05/2024] [Revised: 12/16/2024] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Fusarium circinatum, the causal agent of pine pitch canker, is one of the most destructive pathogens of Pinus species worldwide. Infections by this pathogen result in serious mortality of seedlings due to root and root collar disease, and growth reduction in trees due to canker formation and dieback. Although much is known about the population biology, genetics, and genomics of F. circinatum, relatively little is known regarding the molecular basis of pathogenicity in F. circinatum. In this study, a protoplast-based transformation using CRISPR-Cas9-mediated genome editing was utilized to functionally characterize a putative pathogenicity gene in three different strains of the fungus. In silico analyses suggested the gene likely encodes a small secreted protein, and all isolates in which it was deleted displayed significantly reduced vegetative growth and asexual spore production compared to the wild-type isolates. In pathogenicity tests, lesions induced by the deletion mutants on detached Pinus patula branches were significantly shorter than those produced by the wild-types. The putative pathogenicity gene was named Pgs reflecting its role in pathogenicity, growth, and sporulation. Future research will seek to explore the molecular mechanisms underlying the mutant phenotypes observed. Overall, this study represents a significant advance in F. circinatum research as the development and application of a Cas9-mediated gene deletion process opens new avenues for functional gene characterization underlying many of the pathogen's biological traits.
Collapse
Affiliation(s)
- Alida van Dijk
- Department of Biochemistry, Genetics and Microbiology (BGM), Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa.
| | - Andi M Wilson
- Department of Biochemistry, Genetics and Microbiology (BGM), Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa; Section for Organismal Biology, Department of Plant and Environmental Sciences, University of Copenhagen, Denmark
| | - Bianke Marx
- Department of Biochemistry, Genetics and Microbiology (BGM), Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
| | - Bianca Hough
- Department of Biochemistry, Genetics and Microbiology (BGM), Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
| | - Benedicta Swalarsk-Parry
- Department of Biochemistry, Genetics and Microbiology (BGM), Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
| | - Lieschen De Vos
- Department of Biochemistry, Genetics and Microbiology (BGM), Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
| | - Michael J Wingfield
- Department of Biochemistry, Genetics and Microbiology (BGM), Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
| | - Brenda D Wingfield
- Department of Biochemistry, Genetics and Microbiology (BGM), Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
| | - Emma T Steenkamp
- Department of Biochemistry, Genetics and Microbiology (BGM), Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Pretoria, South Africa
| |
Collapse
|
108
|
Hu Y, Peng Z, Qiu M, Xue L, Ren H, Wu X, Zhu X, Ding Y. Developing biotechnologies in organoids for liver cancer. BIOMEDICAL TECHNOLOGY 2025; 9:100067. [DOI: 10.1016/j.bmt.2024.100067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
109
|
Dederer HG. Human health and genetic technology. Trends Biotechnol 2025; 43:522-532. [PMID: 40015249 DOI: 10.1016/j.tibtech.2024.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/02/2024] [Accepted: 12/16/2024] [Indexed: 03/01/2025]
Abstract
The 1975 Asilomar conference contributed to the misperception that recombinant DNA (rDNA) technology is inherently risky to human health and the environment. It thus paved the way toward process-based regulation of genetically modified organisms (GMOs), such as in the EU. Initially, this regulatory approach obstructed technological uses of rDNA related to human health. However, regulators gradually softened the rules applicable to laboratories or industrial facilities. This encouraged R&D and production of pharmaceuticals derived from GMOs. Nevertheless, administering pharmaceuticals containing GMOs to patients may still be confronted with burdensome process-based GMO law on the deliberate release of GMOs into the environment. On the other hand, pharmaceutical law may need to be updated regarding, for example, novel gene therapies or xenotransplantation.
Collapse
|
110
|
Kadowaki A, Wheeler MA, Li Z, Andersen BM, Lee HG, Illouz T, Lee JH, Ndayisaba A, Zandee SEJ, Basu H, Chao CC, Mahler JV, Klement W, Neel D, Bergstresser M, Rothhammer V, Lipof G, Srun L, Soleimanpour SA, Chiu I, Prat A, Khurana V, Quintana FJ. CLEC16A in astrocytes promotes mitophagy and limits pathology in a multiple sclerosis mouse model. Nat Neurosci 2025; 28:470-486. [PMID: 40033124 PMCID: PMC12039076 DOI: 10.1038/s41593-025-01875-9] [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: 01/09/2023] [Accepted: 12/20/2024] [Indexed: 03/05/2025]
Abstract
Astrocytes promote neuroinflammation and neurodegeneration in multiple sclerosis (MS) through cell-intrinsic activities and their ability to recruit and activate other cell types. In a genome-wide CRISPR-based forward genetic screen investigating regulators of astrocyte proinflammatory responses, we identified the C-type lectin domain-containing 16A gene (CLEC16A), linked to MS susceptibility, as a suppressor of nuclear factor-κB (NF-κB) signaling. Gene and small-molecule perturbation studies in mouse primary and human embryonic stem cell-derived astrocytes in combination with multiomic analyses established that CLEC16A promotes mitophagy, limiting mitochondrial dysfunction and the accumulation of mitochondrial products that activate NF-κB, the NLRP3 inflammasome and gasdermin D. Astrocyte-specific Clec16a inactivation increased NF-κB, NLRP3 and gasdermin D activation in vivo, worsening experimental autoimmune encephalomyelitis, a mouse model of MS. Moreover, we detected disrupted mitophagic capacity and gasdermin D activation in astrocytes in samples from individuals with MS. These findings identify CLEC16A as a suppressor of astrocyte pathological responses and a candidate therapeutic target in MS.
Collapse
MESH Headings
- Animals
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Mitophagy/physiology
- Mitophagy/genetics
- Astrocytes/metabolism
- Astrocytes/pathology
- Mice
- Multiple Sclerosis/pathology
- Multiple Sclerosis/metabolism
- Multiple Sclerosis/genetics
- Humans
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Monosaccharide Transport Proteins/genetics
- Monosaccharide Transport Proteins/metabolism
- Disease Models, Animal
- Mice, Inbred C57BL
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Inflammasomes/metabolism
- Intracellular Signaling Peptides and Proteins/metabolism
- NF-kappa B/metabolism
- Mitochondria/metabolism
- Female
Collapse
Affiliation(s)
- Atsushi Kadowaki
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Immunology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Neurology, Graduate School of Medicine, Faculty of Medicine, The University of Osaka, Suita, Japan
| | - Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zhaorong Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian M Andersen
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, VA Medical Center, Boston, MA, USA
| | - Hong-Gyun Lee
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tomer Illouz
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joon-Hyuk Lee
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alain Ndayisaba
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephanie E J Zandee
- Neuroimmunology Research Laboratory, CRCHUM and Department of Neuroscience, Faculty of Medicine, Universite de Montreal, Montreal, Quebec, Canada
| | - Himanish Basu
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Chun-Cheih Chao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joao V Mahler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wendy Klement
- Neuroimmunology Research Laboratory, CRCHUM and Department of Neuroscience, Faculty of Medicine, Universite de Montreal, Montreal, Quebec, Canada
| | - Dylan Neel
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | | | - Veit Rothhammer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gabriel Lipof
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lena Srun
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Scott A Soleimanpour
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Isaac Chiu
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Alexandre Prat
- Neuroimmunology Research Laboratory, CRCHUM and Department of Neuroscience, Faculty of Medicine, Universite de Montreal, Montreal, Quebec, Canada
| | - Vikram Khurana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
111
|
Salman A, Song WK, Storm T, McClements ME, MacLaren RE. CRISPR targeting of SNPs associated with age-related macular degeneration in ARPE-19 cells: a potential model for manipulating the complement system. Gene Ther 2025; 32:132-141. [PMID: 40102632 PMCID: PMC11946884 DOI: 10.1038/s41434-025-00522-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: 07/17/2024] [Revised: 01/10/2025] [Accepted: 02/14/2025] [Indexed: 03/20/2025]
Abstract
Age-related Macular degeneration (AMD) is a major cause of vision loss and is linked to several predisposing single nucleotide polymorphisms (SNPs). CRISPR-mediated genome editing offers the potential to target negatively associated SNPs in an allele-specific manner, necessitating the need for a relevant cell model. The ARPE-19 cell line, with its stable monolayer growth and retinal pigment epithelium (RPE) characteristics, serves as an ideal model for AMD studies. Chronic inflammation and complement system dysregulation are implicated in AMD pathogenesis. Most genetic variations associated with AMD are in complement genes, suggesting their regulatory role. In this study, we conducted targeted PCRs to identify AMD-related SNPs in ARPE-19 cells and used CRISPR constructs to assess allele-specific activity. Guide RNA sequences were cloned into an EF-1-driven SpCas9 vector and packaged into lentivirus. Targeting efficiencies were evaluated with TIDE analysis, and allele-specificity was measured with NGS analysis 30 days post-transduction. Our results showed varying targeting efficiencies depending on guide RNA efficacy. For example, TIDE analysis of CFH SNPs rs1061170 and rs1410996 revealed efficiencies of 35.5% and 33.8%, respectively. CFB SNP rs4541862 showed efficiencies from 3% to 36.7%, and rs641153 ranged from 3.4% to 23.8%. Additionally, allele-specific targeting of AMD-related SNPs rs1061170, rs1410996, rs4541862, and rs641153 ranged from 48% to 52% in heterozygous differentiated ARPE-19 cells. These findings demonstrate the potential to manipulate the complement system in an AMD model by targeting disease-associated SNPs in an allele-specific manner, offering a promising therapeutic approach.
Collapse
Affiliation(s)
- Ahmed Salman
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| | - Won Kyung Song
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Gangnum Yonsei Eye Clinic, Seoul, Republic of South Korea
| | - Tina Storm
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Robert E MacLaren
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
112
|
Fritz JH, Kufer TA. Editorial: Methods in molecular innate immunity: 2022. Front Immunol 2025; 16:1576957. [PMID: 40092982 PMCID: PMC11906471 DOI: 10.3389/fimmu.2025.1576957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Affiliation(s)
- Jörg H Fritz
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- McGill University Research Center on Complex Traits (MRCCT), McGill University, Montréal, QC, Canada
- Dahdaleh Institute of Genomic Medicine (DIgM), McGill University, Montréal, QC, Canada
| | - Thomas A Kufer
- Department of Immunology, Institute for Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
113
|
Lobo S, Barbosa-Matos R, Dória S, Pedro AM, Brito A, Ferreira D, Oliveira C. A protocol for karyotyping and genetic editing of induced pluripotent stem cells with homology-directed repair mediated CRISPR/Cas9. Biol Methods Protoc 2025; 10:bpaf018. [PMID: 40124668 PMCID: PMC11930342 DOI: 10.1093/biomethods/bpaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/30/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
CRISPR/Cas9-mediated homology-directed repair (HDR) allows precise gene editing, but its efficiency remains low for certain cell types, such as human induced pluripotent stem cells (hiPSCs). In this study, we aimed to introduce the CTNNA1: c.2023C>T (p.Q675*) genetic alteration, which is associated with Hereditary Diffuse Gastric Cancer, into hiPSCs using CRISPR/Cas9. We designed a single-guide RNA targeting the alteration site and a single-stranded oligonucleotide donor DNA template for HDR-based repair. Herein, we report the successful introduction of the CTNNA1: c.2023C>T homozygous alteration in one hiPSC line, which resulted in severe phenotypic changes, including impaired colony formation and cell proliferation. Additionally, we established a straightforward protocol to assess hiPSCs karyotype integrity, ensuring the chromosomal stability required for the gene-editing process. This protocol involves routine G-banding analysis that is required for regular quality controls during handling of hiPSCs. This study demonstrates an efficient approach to precisely edit hiPSCs by CRISPR/Cas9 and highlights the essential role of CTNNA1 expression in maintaining hiPSC viability. Our methodology provides a valuable framework for modeling disease-associated alterations in human-derived cellular models that can be reproduced for other genes and other types of cell lines.
Collapse
Affiliation(s)
- Silvana Lobo
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, 4200-135, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, 4050-313, Portugal
| | - Rita Barbosa-Matos
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, 4200-135, Portugal
| | - Sofia Dória
- Genetics Unit, Department of Pathology, Faculty of Medicine, University of Porto, Porto, 4200-319, Portugal
| | - Ana Maria Pedro
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, 4200-135, Portugal
| | - Ana Brito
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, 4200-135, Portugal
| | - Daniel Ferreira
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, 4200-135, Portugal
| | - Carla Oliveira
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, 4200-135, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, Porto, 4200-135, Portugal
- FMUP—Faculdade de Medicina, University of Porto, Porto, 4200-319, Portugal
| |
Collapse
|
114
|
McAndrew MJ, King MB, Lapinaite A. Preparation of high-purity RNPs of CRISPR-based DNA base editors. Methods Enzymol 2025; 712:277-315. [PMID: 40121077 DOI: 10.1016/bs.mie.2025.01.019] [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]
Abstract
Since their introduction, CRISPR-based DNA base editors (BEs) have become essential in the field of precision genome editing, revolutionizing the correction of pathogenic SNPs for both basic research and therapeutic applications. As this technology advances, more laboratories are implementing these tools into their workflow. The delivery of BEs as BE-guide RNA complexes (RNPs), rather than as mRNA or plasmids, has been shown to exhibit lower off-target effects, establishing it as the preferred method of delivery. However, there are no protocols describing in detail how to obtain high-purity and highly active BE RNPs. Here, we offer a comprehensive guide for the expression, purification, RNP reconstitution, and in vitro activity assessment of TadA-based BEs. The protocol includes guidance on performing activity assays using commercial denaturing gels, which is convenient and uses standard molecular biology equipment. This allows for rapid quality control testing of reconstituted BE RNPs prior to more expensive and time-consuming in vivo genome editing experiments. Overall, this protocol aims to empower more laboratories to generate tailored BE RNPs for diverse in vitro and in vivo applications.
Collapse
Affiliation(s)
- Mitchell J McAndrew
- School of Molecular Sciences, Arizona State University, Tempe, AZ, United States; Gavin Herbert Eye Institute - Center for Translational Vision Research, University of California Irvine, School of Medicine, Irvine, CA, United States
| | - Madeleine B King
- School of Molecular Sciences, Arizona State University, Tempe, AZ, United States; Gavin Herbert Eye Institute - Center for Translational Vision Research, University of California Irvine, School of Medicine, Irvine, CA, United States
| | - Audrone Lapinaite
- Gavin Herbert Eye Institute - Center for Translational Vision Research, University of California Irvine, School of Medicine, Irvine, CA, United States; Department of Ophthalmology, University of California Irvine, School of Medicine, Irvine, CA, United States; Department of Biomedical Engineering, University of California Irvine, Irvine, CA, United States.
| |
Collapse
|
115
|
Manzourolajdad A, Mohebbi M. Secondary-Structure-Informed RNA Inverse Design via Relational Graph Neural Networks. Noncoding RNA 2025; 11:18. [PMID: 40126342 PMCID: PMC11932209 DOI: 10.3390/ncrna11020018] [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: 12/24/2024] [Revised: 01/31/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
RNA inverse design is an essential part of many RNA therapeutic strategies. To date, there have been great advances in computationally driven RNA design. The current machine learning approaches can predict the sequence of an RNA given its 3D structure with acceptable accuracy and at tremendous speed. The design and engineering of RNA regulators such as riboswitches, however, is often more difficult, partly due to their inherent conformational switching abilities. Although recent state-of-the-art models do incorporate information about the multiple structures that a sequence can fold into, there is great room for improvement in modeling structural switching. In this work, a relational geometric graph neural network is proposed that explicitly incorporates alternative structures to predict an RNA sequence. Converting the RNA structure into a geometric graph, the proposed model uses edge types to distinguish between the primary structure, secondary structure, and spatial positioning of the nucleotides in representing structures. The results show higher native sequence recovery rates over those of gRNAde across different test sets (eg. 72% vs. 66%) and a benchmark from the literature (60% vs. 57%). Secondary-structure edge types had a more significant impact on the sequence recovery than the spatial edge types as defined in this work. Overall, these results suggest the need for more complex and case-specific characterization of RNA for successful inverse design.
Collapse
Affiliation(s)
- Amirhossein Manzourolajdad
- Department of Computer Science, State University of New York Polytechnic Institute, 100 Seymour Rd., Utica, NY 13502, USA
| | - Mohammad Mohebbi
- Department of Computer Science and Information Science, University of North Georgia, Dahlonega, GA 30597, USA;
| |
Collapse
|
116
|
Joshi CK, Jamasb AR, Viñas R, Harris C, Mathis SV, Morehead A, Anand R, Liò P. gRNAde: Geometric Deep Learning for 3D RNA inverse design. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.31.587283. [PMID: 38826198 PMCID: PMC11142113 DOI: 10.1101/2024.03.31.587283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Computational RNA design tasks are often posed as inverse problems, where sequences are designed based on adopting a single desired secondary structure without considering 3D conformational diversity. We introduce gRNAde, a geometric RNA design pipeline operating on 3D RNA backbones to design sequences that explicitly account for structure and dynamics. gRNAde uses a multi-state Graph Neural Network and autoregressive decoding to generates candidate RNA sequences conditioned on one or more 3D backbone structures where the identities of the bases are unknown. On a single-state fixed backbone re-design benchmark of 14 RNA structures from the PDB identified by Das et al. (2010), gRNAde obtains higher native sequence recovery rates (56% on average) compared to Rosetta (45% on average), taking under a second to produce designs compared to the reported hours for Rosetta. We further demonstrate the utility of gRNAde on a new benchmark of multi-state design for structurally flexible RNAs, as well as zero-shot ranking of mutational fitness landscapes in a retrospective analysis of a recent ribozyme. Experimental wet lab validation on 10 different structured RNA backbones finds that gRNAde has a success rate of 50% at designing pseudoknotted RNA structures, a significant advance over 35% for Rosetta. Open source code and tutorials are available at: github.com/chaitjo/geometric-rna-design.
Collapse
|
117
|
Matera MG, Calzetta L, Rinaldi B, de Novellis V, Page CP, Barnes PJ, Cazzola M. Animal models of chronic obstructive pulmonary disease and their role in drug discovery and development: a critical review. Expert Opin Drug Discov 2025:1-20. [PMID: 39939153 DOI: 10.1080/17460441.2025.2466704] [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: 11/24/2024] [Revised: 01/16/2025] [Accepted: 02/10/2025] [Indexed: 02/14/2025]
Abstract
INTRODUCTION The use of laboratory animals is essential to understand the mechanisms underlying COPD and to discover and evaluate new drugs. However, the complex changes associated with the disease in humans are difficult to fully replicate in animal models. AREAS COVERED This review examines the most recent literature on animal models of COPD and their implications for drug discovery and development. EXPERT OPINION Recent advances in animal models include the introduction of transgenic mice with an increased propensity to develop COPD-associated features, such as emphysema, and animals exposed to relevant environmental agents other than cigarette smoke, in particular biomass smoke and other air pollutants. Other animal species, including zebrafish, pigs, ferrets and non-human primates, are also increasingly being used to gain insights into human COPD. Furthermore, three-dimensional organoids and humanized mouse models are emerging as technologies for evaluating novel therapeutics in more human-like models. However, despite these advances, no model has yet fully captured the heterogeneity and progression of COPD as observed in humans. Therefore, further research is needed to develop improved models incorporating humanized elements in experimental animals, that may better predict therapeutic responses in clinic settings and accelerate the development of new treatments for this debilitating disease.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Luigino Calzetta
- Department of Medicine and Surgery, Respiratory Disease and Lung Function Unit, University of Parma, Parma, Italy
| | - Barbara Rinaldi
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Vito de Novellis
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Clive P Page
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Peter J Barnes
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|
118
|
Weldemichael MY, Gebremedhn HM. Enhancing tiny millets through genome editing: current status and future prospects. Mol Genet Genomics 2025; 300:22. [PMID: 39982542 DOI: 10.1007/s00438-025-02231-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
This study aims to address the critical need for genetic improvement of small millets, which are vital yet underutilized cereal crops cultivated in semi-arid regions of Africa and Asia. Given their high nutritional value and climate resilience, small millets hold significant potential for food security and sustainable agriculture in arid regions. However, traditional breeding methods have proven to be time-consuming and inefficient in enhancing desirable traits. This study highlights the transformative potential of genome editing technologies, particularly the CRISPR/Cas9 system, in accelerating the development of improved small millet varieties. The findings presented in this paper detail recent advancements in using CRISPR/Cas for enhancing resistance to biotic stresses, including bacterial, viral, and fungal pathogens. Additionally, we explore how genome editing can be applied to improve abiotic stress tolerance, addressing challenges such as drought, cold, heat, and herbicides in small millets. We discuss the existing challenges faced by breeders, including issues related to ploidy levels, off-target effects, and limitations in organelle genome modification. The review also suggests potential strategies for overcoming these bottlenecks, aiming to develop stress-resistant super cultivars. Overall, this paper provides an overview of the current state of genome editing research in small millets while identifying future opportunities to enhance key traits for nutrient enrichment and climate resilience, ultimately paving the way for advancements in these crucial crops.
Collapse
Affiliation(s)
- Micheale Yifter Weldemichael
- Department of Biotechnology, College of Dryland Agriculture and Natural Resources, Mekelle University, P.O. Box 231, Mekelle, Tigrai, Ethiopia.
| | - Hailay Mehari Gebremedhn
- Department of Biotechnology, College of Dryland Agriculture and Natural Resources, Mekelle University, P.O. Box 231, Mekelle, Tigrai, Ethiopia
| |
Collapse
|
119
|
De Paula VS, Dubey A, Arthanari H, Sgourakis NG. Dynamic sampling of a surveillance state enables DNA proofreading by Cas9. Cell Chem Biol 2025; 32:267-279.e5. [PMID: 39471812 PMCID: PMC12051036 DOI: 10.1016/j.chembiol.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/22/2024] [Accepted: 10/03/2024] [Indexed: 11/01/2024]
Abstract
CRISPR-Cas9 has revolutionized genome engineering applications by programming its single-guide RNA, where high specificity is required. However, the precise molecular mechanism underscoring discrimination between on/off-target DNA sequences, relative to the guide RNA template, remains elusive. Here, using methyl-based NMR to study multiple holoenzymes assembled in vitro, we elucidate a discrete protein conformational state which enables recognition of DNA mismatches at the protospacer adjacent motif (PAM)-distal end. Our results delineate an allosteric pathway connecting a dynamic conformational switch at the REC3 domain, with the sampling of a catalytically competent state by the HNH domain. Our NMR data show that HiFi Cas9 (R691A) increases the fidelity of DNA recognition by stabilizing this "surveillance state" for mismatched substrates, shifting the Cas9 conformational equilibrium away from the active state. These results establish a paradigm of substrate recognition through an allosteric protein-based switch, providing unique insights into the molecular mechanism which governs Cas9 selectivity.
Collapse
Affiliation(s)
- Viviane S De Paula
- Center for Computational and Genomic Medicine, The Children's Hospital of Philadelphia, 3501 Civic Center Boulevard, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, PA 19104-6059, USA.
| | - Abhinav Dubey
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Haribabu Arthanari
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nikolaos G Sgourakis
- Center for Computational and Genomic Medicine, The Children's Hospital of Philadelphia, 3501 Civic Center Boulevard, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, PA 19104-6059, USA.
| |
Collapse
|
120
|
Ramírez-Cortés F, Ménová P. Hepatocyte targeting via the asialoglycoprotein receptor. RSC Med Chem 2025; 16:525-544. [PMID: 39628900 PMCID: PMC11609720 DOI: 10.1039/d4md00652f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
This review highlights the potential of asialoglycoprotein receptor (ASGPR)-mediated targeting in advancing liver-specific treatments and underscores the ongoing progress in the field. First, we provide a comprehensive examination of the nature of ASGPR ligands, both natural and synthetic. Next, we explore various drug delivery strategies leveraging ASGPR, with a particular emphasis on the delivery of therapeutic nucleic acids such as small interfering RNAs (siRNAs) and antisense oligonucleotides (ASOs). An in-depth analysis of the current status of RNA interference (RNAi) and ASO-based therapeutics is included, detailing approved therapies and those in various stages of clinical development (phases 1 to 3). Afterwards, we give an overview of other ASGPR-targeted conjugates, such as those with peptide nucleic acids or aptamers. Finally, targeted protein degradation of extracellular proteins through ASGPR is briefly discussed.
Collapse
Affiliation(s)
| | - Petra Ménová
- University of Chemistry and Technology, Prague Technická 5 16628 Prague 6 Czech Republic
| |
Collapse
|
121
|
McMahon M, Maquat LE. Exploring the therapeutic potential of modulating nonsense-mediated mRNA decay. RNA (NEW YORK, N.Y.) 2025; 31:333-348. [PMID: 39667907 PMCID: PMC11874985 DOI: 10.1261/rna.080334.124] [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: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Discovered more than four decades ago, nonsense-mediated mRNA decay (NMD) plays a fundamental role in the regulation of gene expression and is a major contributor to numerous diseases. With advanced technologies, several novel approaches aim to directly circumvent the effects of disease-causing frameshift and nonsense mutations. Additional therapeutics aim to globally dampen the NMD pathway in diseases associated with pathway hyperactivation, one example being Fragile X syndrome. In other cases, therapeutics have been designed to hijack or inhibit the cellular NMD machinery to either activate or obviate transcript-specific NMD by modulating pre-mRNA splicing. Here, we discuss promising approaches employed to regulate NMD for therapeutic purposes and highlight potential challenges in future clinical development. We are optimistic that the future of developing target-specific and global modulators of NMD (inhibitors as well as activators) is bright and will revolutionize the treatment of many genetic disorders, especially those with high unmet medical need.
Collapse
Affiliation(s)
- Mary McMahon
- ReviR Therapeutics, Brisbane, California 94005, USA
| | - Lynne E Maquat
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, USA
- Center for RNA Biology, University of Rochester, Rochester, New York 14642, USA
| |
Collapse
|
122
|
Maximiano M, de Sousa LJ, Feitosa GC, Lopes MEM, Ortega B, Madeiro RDS, Távora FTK, Pereira BM, Brilhante de Oliveira Neto O, Ulhôa CJ, Brasileiro ACM, Aragão FJL, Mehta A, Franco OL. Unlocking Nature's Shield: The Promising Potential of CRISPRa in Amplifying Antimicrobial Peptide Expression in Common Bean ( Phaseolus vulgaris L.). ACS OMEGA 2025; 10:5909-5918. [PMID: 39989781 PMCID: PMC11840611 DOI: 10.1021/acsomega.4c09817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 02/25/2025]
Abstract
This study proposes using the CRISPR transcriptional activation strategy to modulate the expression of genes encoding defense proteins and antimicrobial peptides (AMPs) in Phaseolus vulgaris. Three genes (PvD1, Pv-thionin, and Pv-lectin) were selected and targeted by the CRISPR-dCas9-TV-mediated transcriptional activation complex in the P. vulgaris L. hairy root. RT-qPCR investigated their activation efficiency. The eGFP-positive transgenic hairy roots exhibit enhanced expression of targeted genes compared to that of control roots. A moderate increase of 1.37-fold in PvD1 gene expression was observed in transgenic hairy roots, while 6.97-fold (Pv-lectin) and 5.70-fold (Pv-thionin) increases were observed. Importantly, no off-target effects of sgRNAs were detected, ensuring the precision and safety of the CRISPR-dCas9-TV strategy. The present article is a proof-of-concept study, and it has succeeded in demonstrating the efficiency of the CRISPR-dCas9-TV strategy in modulating the expression of target genes in P. vulgaris, paving the way for an alternative approach to protecting such essential crop plants.
Collapse
Affiliation(s)
- Mariana
Rocha Maximiano
- Universidade
Católica de Brasília, Centro de Análises Proteômicas e Bioquímicas,
Programa de Pós-Graduação em Ciências
Genômicas e Biotecnologia, Brasília CEP: 71966-700, Distrito Federal, Brazil
- Universidade
Católica Dom Bosco, S-Inova Biotech,
Pós-Graduação em Biotecnologia, Campo Grande CEP: 79117-900, Mato Grosso do Sul, Brazil
| | - Lucas José de Sousa
- Universidade
de Brasília, Brasília CEP: 70910-900, Distrito Federal, Brazil
- Embrapa
Recursos Genéticos e Biotecnologia, Brasília CEP: 70770-917, Distrito Federal, Brazil
| | - Gabriel Cidade Feitosa
- Universidade
Católica de Brasília, Centro de Análises Proteômicas e Bioquímicas,
Programa de Pós-Graduação em Ciências
Genômicas e Biotecnologia, Brasília CEP: 71966-700, Distrito Federal, Brazil
- Universidade
Católica Dom Bosco, S-Inova Biotech,
Pós-Graduação em Biotecnologia, Campo Grande CEP: 79117-900, Mato Grosso do Sul, Brazil
- Universidade
de Brasília, Brasília CEP: 70910-900, Distrito Federal, Brazil
| | - Maria Eduarda Melo Lopes
- Universidade
Católica de Brasília, Centro de Análises Proteômicas e Bioquímicas,
Programa de Pós-Graduação em Ciências
Genômicas e Biotecnologia, Brasília CEP: 71966-700, Distrito Federal, Brazil
- Centro
Universitário do Distrito Federal, Brasília CEP: 70390-030, Distrito Federal, Brazil
| | - Brisa Ortega
- Universidade
Católica de Brasília, Centro de Análises Proteômicas e Bioquímicas,
Programa de Pós-Graduação em Ciências
Genômicas e Biotecnologia, Brasília CEP: 71966-700, Distrito Federal, Brazil
- Centro
Universitário do Distrito Federal, Brasília CEP: 70390-030, Distrito Federal, Brazil
| | - Raquel dos Santos Madeiro
- Universidade
Católica de Brasília, Centro de Análises Proteômicas e Bioquímicas,
Programa de Pós-Graduação em Ciências
Genômicas e Biotecnologia, Brasília CEP: 71966-700, Distrito Federal, Brazil
- Centro
Universitário do Distrito Federal, Brasília CEP: 70390-030, Distrito Federal, Brazil
| | | | - Bruna Medeiros Pereira
- Embrapa
Recursos Genéticos e Biotecnologia, Brasília CEP: 70770-917, Distrito Federal, Brazil
| | | | | | | | | | - Angela Mehta
- Embrapa
Recursos Genéticos e Biotecnologia, Brasília CEP: 70770-917, Distrito Federal, Brazil
| | - Octávio Luiz Franco
- Universidade
Católica de Brasília, Centro de Análises Proteômicas e Bioquímicas,
Programa de Pós-Graduação em Ciências
Genômicas e Biotecnologia, Brasília CEP: 71966-700, Distrito Federal, Brazil
- Universidade
Católica Dom Bosco, S-Inova Biotech,
Pós-Graduação em Biotecnologia, Campo Grande CEP: 79117-900, Mato Grosso do Sul, Brazil
| |
Collapse
|
123
|
Correia CD, Calado SM, Matos A, Esteves F, De Sousa-Coelho AL, Campinho MA, Fernandes MT. Advancing Glioblastoma Research with Innovative Brain Organoid-Based Models. Cells 2025; 14:292. [PMID: 39996764 PMCID: PMC11854129 DOI: 10.3390/cells14040292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/06/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
Glioblastoma (GBM) is a relatively rare but highly aggressive form of brain cancer characterized by rapid growth, invasiveness, and resistance to standard therapies. Despite significant progress in understanding its molecular and cellular mechanisms, GBM remains one of the most challenging cancers to treat due to its high heterogeneity and complex tumor microenvironment. To address these obstacles, researchers have employed a range of models, including in vitro cell cultures and in vivo animal models, but these often fail to replicate the complexity of GBM. As a result, there has been a growing focus on refining these models by incorporating human-origin cells, along with advanced genetic techniques and stem cell-based bioengineering approaches. In this context, a variety of GBM models based on brain organoids were developed and confirmed to be clinically relevant and are contributing to the advancement of GBM research at the preclinical level. This review explores the preparation and use of brain organoid-based models to deepen our understanding of GBM biology and to explore novel therapeutic approaches. These innovative models hold significant promise for improving our ability to study this deadly cancer and for advancing the development of more effective treatments.
Collapse
Affiliation(s)
- Cátia D. Correia
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve (UAlg), Campus de Gambelas, 8005-139 Faro, Portugal
| | - Sofia M. Calado
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
- Faculdade de Ciências e Tecnologia (FCT), Universidade dos Açores (UAc), 9500-321 Ponta Delgada, Portugal
| | - Alexandra Matos
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
| | - Filipa Esteves
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve (UAlg), Campus de Gambelas, 8005-139 Faro, Portugal
| | - Ana Luísa De Sousa-Coelho
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
- Escola Superior de Saúde (ESS), Universidade do Algarve (UAlg), Campus de Gambelas, 8005-139 Faro, Portugal
| | - Marco A. Campinho
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve (UAlg), Campus de Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
- Escola Superior de Saúde (ESS), Universidade do Algarve (UAlg), Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
124
|
Vafaeian A, Rajabi F, Rezaei N. Toll-like receptors in atopic dermatitis: pathogenesis and therapeutic implications. Heliyon 2025; 11:e42226. [PMID: 40007792 PMCID: PMC11850170 DOI: 10.1016/j.heliyon.2025.e42226] [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: 05/04/2024] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Toll-like receptors (TLR), the key players of the innate immune system, contribute to the pathogenesis of atopic dermatitis (AD) through multiple pathways. TLRs play a crucial role in delaying barrier repair, promoting Th2-mediated dermatitis, shifting the response toward Th1 in the chronic phase, and contributing to the establishment of the itch-scratch cycle, as well as mediating the effects of UV radiation. The dysregulation of proinflammatory and immunomodulatory effects of TLRs can be attributed to their ligand structures, receptor heterodimerization, the relative frequency of each TLR, interactions with other receptors/signalling pathways, cytokine milieu, and genetic polymorphisms. Current AD treatments like vitamin-D analogs, tacrolimus, and cyclosporine partially work through TLR modulation. Direct TLR stimulation using different compounds has shown therapeutic benefits in preclinical studies. However, significant challenges exist, including off-target effects due to ubiquitous TLR expression and complex roles in immune responses. Future directions include CRISPR-based gene editing to understand TLR functions, development of specific TLR modulators for targeted therapy, and machine learning applications to predict drug responses and identify novel ligands. Patient heterogeneity, including the presence or absence of polymorphisms, variations in TLR expression levels, and differences in immune responses, underscores the need for personalized therapeutic approaches.
Collapse
Affiliation(s)
- Ahmad Vafaeian
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Autoimmune Bullous Diseases Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fateme Rajabi
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Center for Research & Training in Skin Diseases & Leprosy, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Sheffield, UK
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
125
|
Garcia-Montero M, Fanous Y, Krahn AD, Davies B, Cadrin-Tourigny J, Roberts JD. New Insights Into Genetic Right Ventricular Cardiomyopathies. Can J Cardiol 2025:S0828-282X(25)00130-8. [PMID: 39956378 DOI: 10.1016/j.cjca.2025.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/24/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025] Open
Abstract
Inherited right ventricular disease in the form of arrhythmogenic right ventricular cardiomyopathy (ARVC) was first described 40 years ago. The ARVC-causing genes have progressively been identified from the year 2000, accompanied by a robust journey of deep phenotyping. The explosion of genotype and phenotype data coupled with a collaborative spirit in the ARVC community has led to an immense advance in our understanding of the various faces of this disease, with a recent focus on gene-specific phenotypes and risk assessment and mitigation. The modern cardiogenetic team has a wealth of information that informs the biology of the disease, its phenotypic expression, and the processes of care to detect the presence and progression of disease. Gene-specific considerations will raise the bar in precision medicine applied to diagnosis, natural history, and potentially curative interventions with targeted small molecules and gene therapy. This is an exciting time for the ARVC collaborative community to usher in a new era in changing the course of ARVC for patients and their families.
Collapse
Affiliation(s)
- Marta Garcia-Montero
- Cardiovascular Genetics Centre and Electrophysiology Service, Montréal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Yehia Fanous
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - Andrew D Krahn
- Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Brianna Davies
- Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julia Cadrin-Tourigny
- Cardiovascular Genetics Centre and Electrophysiology Service, Montréal Heart Institute, Université de Montréal, Montréal, Québec, Canada
| | - Jason D Roberts
- Section of Cardiac Electrophysiology, Division of Cardiology, Department of Medicine, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada; Population Health Research Institute, Hamilton, Ontario, Canada
| |
Collapse
|
126
|
Bell C, Kilo L, Gottschalk D, Arian J, Deneke L, Kern H, Rickert C, Kobler O, Strauß J, Heine M, Duch C, Ryglewski S. Specific presynaptic functions require distinct Drosophila Ca v2 splice isoforms. eLife 2025; 13:RP100394. [PMID: 39951027 PMCID: PMC11828482 DOI: 10.7554/elife.100394] [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: 02/16/2025] Open
Abstract
At many vertebrate synapses, presynaptic functions are tuned by expression of different Cav2 channels. Most invertebrate genomes contain only one Cav2 gene. The Drosophila Cav2 homolog, cacophony (cac), induces synaptic vesicle release at presynaptic active zones (AZs). We hypothesize that Drosophila cac functional diversity is enhanced by two mutually exclusive exon pairs that are not conserved in vertebrates, one in the voltage sensor and one in the loop binding Caβ and Gβγ subunits. We find that alternative splicing in the voltage sensor affects channel activation voltage. Only the isoform with the higher activation voltage localizes to AZs at the glutamatergic Drosophila larval neuromuscular junction and is imperative for normal synapse function. By contrast, alternative splicing at the other alternative exon pair tunes multiple aspects of presynaptic function. While expression of one exon yields normal transmission, expression of the other reduces channel number in the AZ and thus release probability. This also abolishes presynaptic homeostatic plasticity. Moreover, reduced channel number affects short-term plasticity, which is rescued by increasing the external calcium concentration to match release probability to control. In sum, in Drosophila alternative splicing provides a mechanism to regulate different aspects of presynaptic functions with only one Cav2 gene.
Collapse
Affiliation(s)
- Christopher Bell
- Johannes Gutenberg University Mainz, Institute of Developmental Biology and Neurobiology, Biocenter 1MainzGermany
| | - Lukas Kilo
- RWTH Aachen University, Lehrstuhl für EntwicklungsbiologieAachenGermany
| | - Daniel Gottschalk
- Johannes Gutenberg University Mainz, Institute of Developmental Biology and Neurobiology, Biocenter 1MainzGermany
| | - Jashar Arian
- Johannes Gutenberg University Mainz, Institute of Developmental Biology and Neurobiology, Biocenter 1MainzGermany
| | - Lea Deneke
- Johannes Gutenberg University Mainz, Institute of Developmental Biology and Neurobiology, Biocenter 1MainzGermany
| | - Hanna Kern
- Johannes Gutenberg University Mainz, Institute of Developmental Biology and Neurobiology, Biocenter 1MainzGermany
| | - Christof Rickert
- Johannes Gutenberg University Mainz, Institute of Developmental Biology and Neurobiology, Biocenter 1MainzGermany
| | - Oliver Kobler
- Leibniz Institute for Neurobiology Magdeburg, Combinatorial NeuroImaging Core FacilityMagdeburgGermany
| | - Julia Strauß
- Johannes Gutenberg University Mainz, Institute of Developmental Biology and Neurobiology, Biocenter 1MainzGermany
| | - Martin Heine
- Johannes Gutenberg University Mainz, Institute of Developmental Biology and Neurobiology, Biocenter 1MainzGermany
| | - Carsten Duch
- Johannes Gutenberg University Mainz, Institute of Developmental Biology and Neurobiology, Biocenter 1MainzGermany
| | - Stefanie Ryglewski
- Johannes Gutenberg University Mainz, Institute of Developmental Biology and Neurobiology, Biocenter 1MainzGermany
| |
Collapse
|
127
|
Zhang S, Liu K, Liu Y, Hu X, Gu X. The role and application of bioinformatics techniques and tools in drug discovery. Front Pharmacol 2025; 16:1547131. [PMID: 40017606 PMCID: PMC11865229 DOI: 10.3389/fphar.2025.1547131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
The process of drug discovery and development is both lengthy and intricate, demanding a substantial investment of time and financial resources. Bioinformatics techniques and tools can not only accelerate the identification of drug targets and the screening and refinement of drug candidates, but also facilitate the characterization of side effects and the prediction of drug resistance. High-throughput data from genomics, transcriptomics, proteomics, and metabolomics make significant contributions to mechanics-based drug discovery and drug reuse. This paper summarizes bioinformatics technologies and tools in drug research and development and their roles and applications in drug research and development, aiming to provide references for the development of new drugs and the realization of precision medicine.
Collapse
Affiliation(s)
- Shujun Zhang
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Kaijie Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yafeng Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Xinjun Hu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
- Henan Medical Key Laboratory of Gastrointestinal Microecology and Hepatology, Luoyang, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| |
Collapse
|
128
|
Chey YCJ, Gierus L, Lushington C, Arudkumar JC, B Geiger A, Staker LG, Robertson LJ, Pfitzner C, Kennedy JG, Lee RHB, Godahewa GI, Adikusuma F, Thomas PQ. Optimal SpCas9- and SaCas9-mediated gene editing by enhancing gRNA transcript levels through scaffold poly-T tract reduction. BMC Genomics 2025; 26:138. [PMID: 39939860 PMCID: PMC11823040 DOI: 10.1186/s12864-025-11317-2] [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: 10/08/2024] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
Ensuring sufficient gRNA transcript levels is critical for obtaining optimal CRISPR-Cas9 gene editing efficiency. The standard gRNA scaffold contains a sequence of four thymine nucleotides (4T), which is known to inhibit transcription from Pol III promoters such as the U6 promoter. Our study showed that using standard plasmid transfection protocols, the presence of these 4Ts did not significantly affect editing efficiency, as most of the gRNAs tested (55 gRNAs) achieved near-perfect editing outcomes. We observed that gRNAs with lower activity were T-rich and had reduced gRNA transcript levels. However, this issue can be effectively resolved by increasing transcript levels, which can be readily achieved by shortening the 4T sequences. In this study, we demonstrated this by modifying the sequences to 3TC. Although the 3TC scaffold modification did not improve editing efficiency for already efficient gRNAs when high vector quantities were available, it proved highly beneficial under conditions of limited vector availability, where the 3TC scaffold yielded higher editing efficiency. Additionally, we demonstrated that the 3TC scaffold is compatible with SpCas9 high-fidelity variants and ABEmax base editing, enhancing their editing efficiency. Another commonly used natural Cas9 variant, SaCas9, also benefited from the 3TC scaffold sequence modification, which increased gRNA transcription and subsequently improved editing activity. This modification was applied to the EDIT-101 therapeutic strategy, where it demonstrated marked improvements in performance. This study highlights the importance of shortening the 4T sequences in the gRNA scaffold to optimize gRNA transcript expression for enhanced CRISPR-Cas9 gene editing efficiency. This optimization is particularly important for therapeutic applications, where the quantity of vector is often limited, ensuring more effective and optimal outcomes.
Collapse
Affiliation(s)
- Yu C J Chey
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Luke Gierus
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Caleb Lushington
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Jayshen C Arudkumar
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Ashleigh B Geiger
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Lachlan G Staker
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Louise J Robertson
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Chandran Pfitzner
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Jesse G Kennedy
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Ryan H B Lee
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Gelshan I Godahewa
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Fatwa Adikusuma
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia.
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.
| | - Paul Q Thomas
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia.
- South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.
- South Australian Genome Editing (SAGE) Facility, SAHMRI, Adelaide, South Australia, Australia.
| |
Collapse
|
129
|
Zhang Y, Ding X, Yang Z, Wang J, Li C, Zhou G. Emerging Microfluidic Building Blocks for Cultured Meat Construction. ACS APPLIED MATERIALS & INTERFACES 2025; 17:8771-8793. [PMID: 39884858 DOI: 10.1021/acsami.4c19276] [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: 02/01/2025]
Abstract
Cultured meat aims to produce meat mass by culturing cells and tissues based on the muscle regeneration mechanism, and is considered an alternative to raising and slaughtering livestock. Hydrogel building blocks are commonly used as substrates for cell culture in tissue engineering and cultured meat because of their high water content, biocompatibility, and similar three-dimensional (3D) environment to the cellular niche in vivo. With the characteristics of precise manipulation of fluids, microfluidics exhibits advantages in the fabrication of building blocks with different structures and components, which have been widely applied in tissue regeneration. Microfluidic building blocks show promising prospects in the field of cultured meat; however, few reviews on the application of microfluidic building blocks in cultured meat have been published. This review outlines the recent status and prospects of the use of microfluidic building blocks in cultured meat. Starting with the introduction of cells and materials for cultured meat tissue construction, we then describe the diverse structures of the fabricated building blocks, including microspheres, microfibers, and microsphere-microfiber hybrid systems. Next, the stacking strategies for tissue construction are highlighted in detail. Finally, challenges and future prospects for developing microfluidic building blocks for cultured meat are discussed.
Collapse
Affiliation(s)
- Yue Zhang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Key Laboratory of Meat Processing, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xi Ding
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Key Laboratory of Meat Processing, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zijiang Yang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Key Laboratory of Meat Processing, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Jie Wang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Key Laboratory of Meat Processing, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chunbao Li
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Key Laboratory of Meat Processing, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Guanghong Zhou
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Key Laboratory of Meat Processing, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
130
|
D’Amico M, Maggiolini FAM, Forleo LR, Cardone MF, Velasco R, Basile T, Bergamini C. Simplified Protocol for the Purification of Native Cas Nucleases for DNA-Free Genome Editing. Methods Protoc 2025; 8:16. [PMID: 39997640 PMCID: PMC11857876 DOI: 10.3390/mps8010016] [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: 01/08/2025] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
DNA-free genome editing by the direct delivery of CRISPR-associated nucleases has emerged as a promising technology due to its precision and reduced risk of off-target effects. However, existing purification protocols for native Cas proteins require the use of complex instrumentation, which limits their application. Here, we present a simplified protocol for the purification of native Cas9, Cas12RR and dCas9-VP64 nucleases optimized for DNA-free genome editing. Our approach leverages a streamlined affinity and ion exchange chromatography coupled with minimal downstream processing, ensuring a good yield and activity of the purified proteins. The in vitro analysis of the purified ribonucleoprotein complex demonstrated a good efficiency of DNA target cleavage. This simplified protocol increases the opportunity to adopt CRISPR technology, and enables broader access to DNA-free genome editing tools also for laboratories that are not specifically equipped for protein purification.
Collapse
Affiliation(s)
- Margherita D’Amico
- CREA Council for Agricultural Research and Economics—Research Center for Viticulture and Enology, Via Casamassima 148, 70010 Turi, Italy; (F.A.M.M.); (L.R.F.); (M.F.C.); (R.V.); (T.B.)
| | | | | | | | | | | | - Carlo Bergamini
- CREA Council for Agricultural Research and Economics—Research Center for Viticulture and Enology, Via Casamassima 148, 70010 Turi, Italy; (F.A.M.M.); (L.R.F.); (M.F.C.); (R.V.); (T.B.)
| |
Collapse
|
131
|
Chepurna O, Chatterjee A, Li Y, Ding H, Murali R, Black KL, Sun T. Nano-Polymers as Cas9 Inhibitors. Polymers (Basel) 2025; 17:417. [PMID: 39940619 PMCID: PMC11820846 DOI: 10.3390/polym17030417] [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] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Despite wide applications of CRISPR/Cas9 technology, effective approaches for CRISPR delivery with functional control are limited. In an attempt to develop a nanoscale CRSIPR/Cas9 delivery platform, we discovered that several biocompatible polymers, including polymalic acid (PMLA), polyglutamic acid (PGA), and polyaspartic acid (PLD), when conjugated with a trileucine (LLL) moiety, can effectively inhibit Cas9 nuclease function. The Cas9 inhibition by those polymers is dose-dependent, with varying efficiency to achieve 100% inhibition. Further biophysical studies revealed that PMLA-LLL directly binds the Cas9 protein, resulting in a substantial decrease in Cas9/sgRNA binding affinity. Transmission electron microscopy and molecular docking were performed to provide a possible binding mechanism for PMLA-LLL to interact with Cas9. This work identified a new class of Cas9 inhibitor in nano-polymer form. These biodegradable polymers may serve as novel Cas9 delivery vehicles with a potential to enhance the precision of Cas9-mediated gene editing.
Collapse
Affiliation(s)
- Oksana Chepurna
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Avradip Chatterjee
- Department of Biomedical Sciences, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yuanqing Li
- Shanghai Institute of Material Medica, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai 200231, China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hong Ding
- Shanghai Institute of Material Medica, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai 200231, China
| | - Ramachandran Murali
- Department of Biomedical Sciences, Research Division of Immunology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Keith L. Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Tao Sun
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
132
|
Abbasi AF, Asim MN, Dengel A. Transitioning from wet lab to artificial intelligence: a systematic review of AI predictors in CRISPR. J Transl Med 2025; 23:153. [PMID: 39905452 PMCID: PMC11796103 DOI: 10.1186/s12967-024-06013-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: 08/27/2024] [Accepted: 12/18/2024] [Indexed: 02/06/2025] Open
Abstract
The revolutionary CRISPR-Cas9 system leverages a programmable guide RNA (gRNA) and Cas9 proteins to precisely cleave problematic regions within DNA sequences. This groundbreaking technology holds immense potential for the development of targeted therapies for a wide range of diseases, including cancers, genetic disorders, and hereditary diseases. CRISPR-Cas9 based genome editing is a multi-step process such as designing a precise gRNA, selecting the appropriate Cas protein, and thoroughly evaluating both on-target and off-target activity of the Cas9-gRNA complex. To ensure the accuracy and effectiveness of CRISPR-Cas9 system, after the targeted DNA cleavage, the process requires careful analysis of the resultant outcomes such as indels and deletions. Following the success of artificial intelligence (AI) in various fields, researchers are now leveraging AI algorithms to catalyze and optimize the multi-step process of CRISPR-Cas9 system. To achieve this goal AI-driven applications are being integrated into each step, but existing AI predictors have limited performance and many steps still rely on expensive and time-consuming wet-lab experiments. The primary reason behind low performance of AI predictors is the gap between CRISPR and AI fields. Effective integration of AI into multi-step CRISPR-Cas9 system demands comprehensive knowledge of both domains. This paper bridges the knowledge gap between AI and CRISPR-Cas9 research. It offers a unique platform for AI researchers to grasp deep understanding of the biological foundations behind each step in the CRISPR-Cas9 multi-step process. Furthermore, it provides details of 80 available CRISPR-Cas9 system-related datasets that can be utilized to develop AI-driven applications. Within the landscape of AI predictors in CRISPR-Cas9 multi-step process, it provides insights of representation learning methods, machine and deep learning methods trends, and performance values of existing 50 predictive pipelines. In the context of representation learning methods and classifiers/regressors, a thorough analysis of existing predictive pipelines is utilized for recommendations to develop more robust and precise predictive pipelines.
Collapse
Affiliation(s)
- Ahtisham Fazeel Abbasi
- Smart Data and Knowledge Services, German Research Center for Artificial Intelligence, 67663, Kaiserslautern, Germany.
- Department of Computer Science, Rhineland-Palatinate Technical University Kaiserslautern-Landau, 67663, Kaiserslautern, Germany.
| | - Muhammad Nabeel Asim
- Department of Computer Science, Rhineland-Palatinate Technical University Kaiserslautern-Landau, 67663, Kaiserslautern, Germany
| | - Andreas Dengel
- Smart Data and Knowledge Services, German Research Center for Artificial Intelligence, 67663, Kaiserslautern, Germany
- Department of Computer Science, Rhineland-Palatinate Technical University Kaiserslautern-Landau, 67663, Kaiserslautern, Germany
| |
Collapse
|
133
|
Zhao Y, Liu D, Yang W, He W, Yan J, Yao L. Resetting the Hsc70-mediated lysosomal degradation of PD-L1 via a supramolecular meso peptide for the restoration of acquired anti-tumor T cell immunity. J Nanobiotechnology 2025; 23:79. [PMID: 39905428 PMCID: PMC11792358 DOI: 10.1186/s12951-025-03171-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/27/2025] [Indexed: 02/06/2025] Open
Abstract
The reduction of cellular PD-L1 abundance through lysosomal degradation is recognized as essential for effective and sustained targeting of PD-L1-dependent immune evasion in cancer. While Hsc70 can interact with PD-L1 to promote its lysosomal degradation, the overexpression of CMTM6 competitively inhibits this interaction, leading to the blockade of PD-L1 lysosomal degradation. To overcome this issue, a meso chimeric peptide PEPPDL1 was designed to specifically bind the PD-1 binding domain of PD-L1 instead of the Hsc70/CMTM6 binding domain, while also binding to Hsc70 to facilitate the dragging of PD-L1 into Hsc70-mediated chaperone-mediated autophagy (CMA), thereby achieving lysosomal degradation. In order to enable internalization into tumor cells, supramolecular engineering techniques were employed through terminal modification involving sulfydryl and monovalent gold ion (Au(I)), both facilitating self-assembly of modified PEPPDL1 into supramolecular nanospheres termed CTAC-PDL1 driven by aurophilic interaction. Furthermore, based on bioinformatics analysis of mRNA expression data from 30 types of tumors obtained from TCGA database, malignant melanoma was identified as the most suitable indication for CTAC-PDL1 due to its specific characteristics of tumor immune. As expected, CTAC-PDL1 effectively reactivated Hsc70-mediated lysosomal degradation of PD-L1 and consequently restored anti-tumor T cell immunity in a B16F10-derived mouse model of malignant melanoma while maintaining a favorable safety profile. Overall, this work not only presents an alternative approach for targeting PD-L1-dependent cancer immune evasion, but also provides a modularized strategy for discovering specific regulators for target proteins in various diseases.
Collapse
Affiliation(s)
- Yujia Zhao
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Dan Liu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Wenguang Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Wangxiao He
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China.
| | - Jin Yan
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Xi'an, China.
| | - Leiqing Yao
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Xi'an, China.
| |
Collapse
|
134
|
Szabó V, Varsányi B, Barboni M, Takács Á, Knézy K, Molnár MJ, Nagy ZZ, György B, Rivolta C. Insights into eye genetics and recent advances in ocular gene therapy. Mol Cell Probes 2025; 79:102008. [PMID: 39805344 DOI: 10.1016/j.mcp.2025.102008] [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: 12/18/2024] [Revised: 01/04/2025] [Accepted: 01/05/2025] [Indexed: 01/16/2025]
Abstract
The rapid advancements in the field of genetics have significantly propelled the development of gene therapies, paving the way for innovative treatments of various hereditary disorders. This review focuses on the genetics of ophthalmologic conditions, highlighting the currently approved ophthalmic gene therapy and exploring emerging therapeutic strategies under development. Inherited retinal dystrophies represent a heterogeneous group of genetic disorders that manifest across a broad spectrum from infancy to late middle age. Key clinical features include nyctalopia (night blindness), constriction of the visual field, impairments in color perception, reduced central visual acuity, and rapid eye movements. Recent technological advancements, such as multimodal imaging, psychophysical assessments, and electrophysiological testing, have greatly enhanced our ability to understand disease progression and establish genotype-phenotype correlations. Additionally, the integration of molecular diagnostics into clinical practice is revolutionizing patient stratification and the design of targeted interventions, underscoring the transformative potential of personalized medicine in ophthalmology. The review also covers the challenges and opportunities in developing gene therapies for other ophthalmic conditions, such as age-related macular degeneration and optic neuropathies. We discuss the viral and non-viral vector systems used in ocular gene therapy, highlighting their advantages and limitations. Additionally, we explore the potential of emerging technologies like CRISPR/Cas9 in treating genetic eye diseases. We briefly address the regulatory landscape, concerns, challenges, and future directions of gene therapy in ophthalmology. We emphasize the need for long-term safety and efficacy data as these innovative treatments move from bench to bedside.
Collapse
Affiliation(s)
- Viktória Szabó
- Semmelweis University, Department of Ophthalmology, Mária Str. 39, Budapest, 1085, Hungary.
| | - Balázs Varsányi
- Semmelweis University, Department of Ophthalmology, Mária Str. 39, Budapest, 1085, Hungary; Ganglion Medical Center, Váradi Str. 10/A, Pécs, 7621, Hungary.
| | - Mirella Barboni
- Semmelweis University, Department of Ophthalmology, Mária Str. 39, Budapest, 1085, Hungary; Institute of Molecular and Clinical Ophthalmology Basel, Mittlere Strasse 91, Basel, CH-4031, Switzerland.
| | - Ágnes Takács
- Semmelweis University, Department of Ophthalmology, Mária Str. 39, Budapest, 1085, Hungary.
| | - Krisztina Knézy
- Semmelweis University, Department of Ophthalmology, Mária Str. 39, Budapest, 1085, Hungary.
| | - Mária Judit Molnár
- Semmelweis University, Institute of Genomic Medicine and Rare Disorders, Gyulai Pál Str. 2, Budapest, 1085, Hungary.
| | - Zoltán Zsolt Nagy
- Semmelweis University, Department of Ophthalmology, Mária Str. 39, Budapest, 1085, Hungary.
| | - Bence György
- Institute of Molecular and Clinical Ophthalmology Basel, Mittlere Strasse 91, Basel, CH-4031, Switzerland; Department of Ophthalmology, University of Basel, Mittlere Strasse 91, Basel, CH-4031, Switzerland.
| | - Carlo Rivolta
- Institute of Molecular and Clinical Ophthalmology Basel, Mittlere Strasse 91, Basel, CH-4031, Switzerland.
| |
Collapse
|
135
|
Lafi Z, Ata T, Asha S. CRISPR in clinical diagnostics: bridging the gap between research and practice. Bioanalysis 2025; 17:281-290. [PMID: 39902531 PMCID: PMC11866644 DOI: 10.1080/17576180.2025.2459520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/24/2025] [Indexed: 02/05/2025] Open
Abstract
CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) has transformed molecular biology through its precise gene-editing capabilities. Beyond its initial applications in genetic modification, CRISPR has emerged as a powerful tool in diagnostics and biosensing. This review explores its transition from genome editing to innovative detection methods, including nucleic acid identification, single nucleotide polymorphism (SNP) analysis, and protein sensing. Advanced technologies such as SHERLOCK and DETECTR demonstrate CRISPR's potential for point-of-care diagnostics, enabling rapid and highly sensitive detection. The integration of chemical modifications, CRISPR-Chip technology, and enzymatic systems like Cas12a and Cas13a enhances signal amplification and detection efficiency. These advancements promise decentralized, real-time diagnostic solutions with significant implications for global healthcare. Furthermore, the fusion of CRISPR with artificial intelligence and digital health platforms is paving the way for more accessible, cost-effective, and scalable diagnostic approaches, ultimately revolutionizing precision medicine.
Collapse
Affiliation(s)
- Zainab Lafi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Tha’er Ata
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Sherine Asha
- School of Medicine, University of Jordan, Amman, Jordan
| |
Collapse
|
136
|
Wei Z, Wang C, Zhang X, Lv Y, Li Y, Gao P, Yang X. CRISPR/Cas9-mediated knockout of Tektin 4-like gene (TEKT4L) causes male sterility of Cydia pomonella. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 177:104257. [PMID: 39756499 DOI: 10.1016/j.ibmb.2025.104257] [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: 09/16/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
The sterile insect technique (SIT) is a well-established and environmentally benign method for population control. Identifying genes that regulate insect fertility while preserving growth and development is crucial for implementing a novel SIT-based pest management approach utilizing CRISPR/Cas9 to target these genes for genetic manipulation. Tektin (TEKT), an essential alpha-helical protein pivotal in sperm formation due to its role in cilia and flagella assembly, has garnered attention. In this study, we identified 7 TEKT genes in the testis of Cydia pomonella, a globally invasive fruit pest. Notably, Tektin4-like (TEKT4L) displayed the highest expression level in male adult especially the testes, suggesting its significance in reproductive processes. By utilizing CRISPR/Cas9 technology to knockout TEKT4L, male sterility was induced, showcasing dominant inherited. When wild-type (WT) females mated with TEKT4L-/- males, eggs laying proceeded normally, but the hatching rate was dramatically reduced, with only 15.49% progressing to the eyespot stage and 68.86% failing to develop normally. The reproductive fitness of TEKT4L-/- males was robust enough to facilitate the transmission of genetic modifications efficiently within the C.pomonella population, yielding a small number of viable offspring. Subsequent cage trials demonstrated the effectiveness of this population in suppressing laboratory populations of C.pomonella, achieving notable results with a relatively low release ratio (TEKT4L-/-♂: WT♂: WT♀ = 5:1:5). Consequently, the targeted disruption of the TEKT4L gene holds promise as a fundamental element in a novel pest control strategy against C. pomonella.
Collapse
Affiliation(s)
- Zihan Wei
- College of Plant Protection, Shenyang Agricultural University, Shenyang, 110866, Liaoning, China; Key Laboratory of Economical and Applied Entomology of Liaoning Province, China; Key Laboratory of Major Agricultural Invasion Biological Monitoring and Control, Shenyang, 110866, Liaoning, China
| | - Chang Wang
- College of Plant Protection, Shenyang Agricultural University, Shenyang, 110866, Liaoning, China; Key Laboratory of Economical and Applied Entomology of Liaoning Province, China; Key Laboratory of Major Agricultural Invasion Biological Monitoring and Control, Shenyang, 110866, Liaoning, China
| | - Xinyue Zhang
- College of Plant Protection, Shenyang Agricultural University, Shenyang, 110866, Liaoning, China; Key Laboratory of Economical and Applied Entomology of Liaoning Province, China; Key Laboratory of Major Agricultural Invasion Biological Monitoring and Control, Shenyang, 110866, Liaoning, China
| | - Yuntong Lv
- College of Plant Protection, Shenyang Agricultural University, Shenyang, 110866, Liaoning, China; Key Laboratory of Economical and Applied Entomology of Liaoning Province, China; Key Laboratory of Major Agricultural Invasion Biological Monitoring and Control, Shenyang, 110866, Liaoning, China
| | - Yuting Li
- College of Plant Protection, Shenyang Agricultural University, Shenyang, 110866, Liaoning, China; Key Laboratory of Economical and Applied Entomology of Liaoning Province, China; Key Laboratory of Major Agricultural Invasion Biological Monitoring and Control, Shenyang, 110866, Liaoning, China
| | - Ping Gao
- College of Plant Protection, Shenyang Agricultural University, Shenyang, 110866, Liaoning, China; Key Laboratory of Economical and Applied Entomology of Liaoning Province, China; Key Laboratory of Major Agricultural Invasion Biological Monitoring and Control, Shenyang, 110866, Liaoning, China
| | - Xueqing Yang
- College of Plant Protection, Shenyang Agricultural University, Shenyang, 110866, Liaoning, China; Key Laboratory of Economical and Applied Entomology of Liaoning Province, China; Key Laboratory of Major Agricultural Invasion Biological Monitoring and Control, Shenyang, 110866, Liaoning, China.
| |
Collapse
|
137
|
Dakal TC, Philip RR, Bhushan R, Sonar PV, Rajagopal S, Kumar A. Genetic and epigenetic regulation of non-coding RNAs: Implications in cancer metastasis, stemness and drug resistance. Pathol Res Pract 2025; 266:155728. [PMID: 39657397 DOI: 10.1016/j.prp.2024.155728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 12/12/2024]
Abstract
Cancer stem cells (CSCs) have a crucial function in the initiation, advancement, and resistance to therapy of tumors. Recent findings indicate that non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), play a complex role in controlling the features of cancer stem cells (CSCs). Non-coding RNAs (ncRNAs) play a crucial role in controlling important characteristics of stem cells, such as their ability to renew themselves, differentiate into distinct cell types, and resist therapy. This article provides an overview of the current understanding of the complex relationship between non-coding RNAs (ncRNAs), namely microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), and cancer stem cells (CSCs). Particular microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are involved in regulating important signaling pathways like as Wnt, Notch, and Hedgehog, which control stem cell-like characteristics. The miR-34, miR-200, and let-7 families specifically aim at inhibiting the process of self-renewal and epithelial-to-mesenchymal transition. On the other hand, long non-coding RNAs (lncRNAs) such as H19, HOTAIR, and MALAT1 play a role in modifying the epigenetic landscape, hence enhancing the characteristics of stemness. This article also offers a thorough examination of the role of non-coding RNAs (ncRNAs) in regulating cancer stemness, emphasizing their impact on crucial biochemical pathways, epigenetic changes, and therapeutic implications. Comprehending the interaction between non-coding RNAs (ncRNAs) and cancer stem cells (CSCs) provides fresh perspectives on possible focused treatments for fighting aggressive and resistant malignancies. Gaining a comprehensive understanding of the connection between non-coding RNA (ncRNA) and cancer stem cells (CSC) offers valuable insights for the development of novel and precise treatments to combat aggressive cancers that are resistant to conventional therapies. In addition, the combination of ncRNA therapies with conventional methods like as chemotherapy or epigenetic medicines could result in synergistic effects. Nevertheless, there are still obstacles to overcome in terms of delivery, effectiveness, and safety. In summary, the interaction between non-coding RNA and cancer stemness shows potential as a targeted treatment approach in the field of precision oncology. This calls for additional investigation and use in clinical settings.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001, India.
| | - Reya Rene Philip
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai, India
| | - Ravi Bhushan
- Department of Zoology, M.S. College, Motihari, Bihar 845401, India
| | | | - Senthilkumar Rajagopal
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka, India
| | - Abhishek Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India; Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India.
| |
Collapse
|
138
|
Arshad S, Qadir ML, Hussain N, Ali Q, Han S, Ali D. Advances in CRISPR/Cas9 technology: shaping the future of photosynthetic microorganisms for biofuel production. FUNCTIONAL PLANT BIOLOGY : FPB 2025; 52:FP24255. [PMID: 39932844 DOI: 10.1071/fp24255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/15/2025] [Indexed: 02/13/2025]
Abstract
Use of fossil fuels causes environmental issues due to its inefficiency and and imminent depletion. This has led to interest in identifying alternative and renewable energy sources such as biofuel generation from photosynthetic organisms. A wide variety of prokaryotic and eukaryotic microorganisms, known as microalgae, have the potential to be economical and ecologically sustainable in the manufacture of biofuels such as bio-hydrogen, biodiesel, bio-oils, and bio-syngas. By using contemporary bioengineering techniques, the innate potential of algae to produce biomass of superior quality may be enhanced. In algal biotechnology, directed genome modification via RNA-guided endonucleases is a new approach. CRISPR/Cas systems have recently been frequently used to modify the genetic makeup of several aquatic and freshwater microalgae. The majority of research has used the Cas9-driven Type II system, one of two classes and six unique kinds of CRISPR systems, to specifically target desired genes in algae, and knock them out and down, or both. Using CRISPR technology to modify its genetic makeup, microalgae has produced more biomass and increased in lipid content. This review highlights the attempts made so far to target microalgae genome modification, discusses the prospects for developing the CRISPR platform for large-scale genome modification of microalgae, and identifies the opportunities and challenges in the development and distribution of CRISPR/Cas9 components.
Collapse
Affiliation(s)
- Samreen Arshad
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Muhammad Luqman Qadir
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Nazim Hussain
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Qurban Ali
- Department of Plant Breeding and Genetics, Faculty of Agricultural Sciences, University of the Punjab, Lahore, Pakistan
| | - Shiming Han
- School of Biological Sciences and Technology, Liupanshui Normal University, Liupanshui 553004, China
| | - Daoud Ali
- Department of Zoology, College of Science, King Saud University, PO Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
139
|
Korody ML, Hildebrandt TB. Progress Toward Genetic Rescue of the Northern White Rhinoceros ( Ceratotherium simum cottoni). Annu Rev Anim Biosci 2025; 13:483-505. [PMID: 39531386 DOI: 10.1146/annurev-animal-111523-102158] [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: 11/16/2024]
Abstract
The northern white rhinoceros (NWR) is functionally extinct, with only two nonreproductive females remaining. However, because of the foresight of scientists, cryopreserved cells and reproductive tissues may aid in the recovery of this species. An ambitious program of natural and artificial gametes and in vitro embryo generation was first outlined in 2015, and many of the proposed steps have been achieved. Multiple induced pluripotent stem cell lines have been established, primordial germ cell-like cells have been generated, oocytes have been collected from the remaining females, blastocysts have been cryopreserved, and the closely related southern white rhinoceros (SWR) is being established as a surrogate. Recently, the first successful embryo transfer in SWR demonstrated that embryos can be generated by in vitro fertilization and cryopreserved. We explore progress to date in using advanced cellular technologies to save the NWR and highlight the necessary next steps to ensure a viable population for reintroduction. We roll out a holistic rescue approach for a charismatic megavertebrate that includes the most advanced cellular technologies, which can provide a blueprint for other critically endangered mammals. We also provide a detailed discussion of the remaining questions in such an upgraded conservation program.
Collapse
Affiliation(s)
- Marisa L Korody
- San Diego Zoo Wildlife Alliance, Escondido, California, USA;
| | - Thomas B Hildebrandt
- Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
- Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany;
| |
Collapse
|
140
|
Yadav S, Prasannan A, Venkatachalam K, Binesh A. Exploring the mechanism and crosstalk between IL-6 and IL- 1β on M2 macrophages under metabolic stress conditions. Cytokine 2025; 186:156852. [PMID: 39765025 DOI: 10.1016/j.cyto.2024.156852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/29/2024] [Accepted: 12/31/2024] [Indexed: 01/14/2025]
Abstract
Macrophages are highly variable immune cells that are important in controlling inflammation and maintaining tissue balance. The ability to polarize into two major types-M1, promoting inflammation, and M2, resolving inflammation and contributing to tissue repair-determines their specific roles in health and disease. M2 macrophages are particularly important for reducing inflammation and promoting tissue regeneration, but their function is shaped mainly by surrounding cells. This is evident in obesity, diabetes, and chronic inflammation. Although many cytokines regulate macrophage polarization, interleukin-6 (IL-6) and interleukin-1β (IL-1β) are major players, but their effects on M2 macrophage behavior under metabolic stress remain unclear. This study describes the intricacies within M2 macrophages concerning IL-6 and IL-1β signaling when under metabolic stress. Though, more frequently than not, IL-6 is labelled as pro-inflammatory, it can also behave as an anti-inflammatory mediator. On the other hand, IL-1β is the main pro-inflammatory agent, particularly in metabolic disorders. The relationship between these cytokines and the macrophages is mediated through important pathways such as JAK/STAT and NFκB, which get perturbed by metabolic stress. Therefore, metabolic stress also alters the functional parameters of macrophages, including alterations in mitochondrial metabolism, glycolytic and oxidative metabolism. Phosphorylation alters the kinetics involved in energy consumption and affects their polarization and their function. However, it has been suggested that IL-6 and IL-1β may work in concert or competition when inducing M2 polarization and, importantly, implicate cytokine release, phagocytic activity, and tissue repair processes. In this review, we discuss the recent literature on the participation of IL-6 and IL-1β cytokines in macrophage polarization and how metabolic stress changes cytokine functions and synergistic relations. A better understanding of these cytokines would serve as an important step toward exploring alternative antiviral strategies directed against metabolic disturbance and, hence, approve further endeavors.
Collapse
Affiliation(s)
- Shawna Yadav
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai 603103, Tamil Nadu, India
| | - Anusha Prasannan
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai 603103, Tamil Nadu, India
| | - Kaliyamurthi Venkatachalam
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai 603103, Tamil Nadu, India
| | - Ambika Binesh
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai 603103, Tamil Nadu, India.
| |
Collapse
|
141
|
Qie B, Tuo J, Chen F, Ding H, Lyu L. Gene therapy for genetic diseases: challenges and future directions. MedComm (Beijing) 2025; 6:e70091. [PMID: 39949979 PMCID: PMC11822459 DOI: 10.1002/mco2.70091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Genetic diseases constitute the majority of rare human diseases, resulting from abnormalities in an individual's genetic composition. Traditional treatments offer limited relief for these challenging conditions. In contrast, the rapid advancement of gene therapy presents significant advantages by directly addressing the underlying causes of genetic diseases, thereby providing the potential for precision treatment and the possibility of curing these disorders. This review aims to delineate the mechanisms and outcomes of current gene therapy approaches in clinical applications across various genetic diseases affecting different body systems. Additionally, genetic muscular disorders will be examined as a case study to investigate innovative strategies of novel therapeutic approaches, including gene replacement, gene suppression, gene supplementation, and gene editing, along with their associated advantages and limitations at both clinical and preclinical levels. Finally, this review emphasizes the existing challenges of gene therapy, such as vector packaging limitations, immunotoxicity, therapy specificity, and the subcellular localization and immunogenicity of therapeutic cargos, while discussing potential optimization directions for future research. Achieving delivery specificity, as well as long-term effectiveness and safety, will be crucial for the future development of gene therapies targeting genetic diseases.
Collapse
Affiliation(s)
- Beibei Qie
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Jianghua Tuo
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Feilong Chen
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Haili Ding
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| | - Lei Lyu
- Institute of Sports Medicine and Health, School of Sports Medicine and HealthChengdu Sport UniversityChengduChina
| |
Collapse
|
142
|
Liang J, Xiao F, Ojo J, Chao WH, Ahmad B, Alam A, Abbas S, Abdelhafez MM, Rahman N, Khan KA, Ghramh HA, Ali J, Chen R. Insect Resistance to Insecticides: Causes, Mechanisms, and Exploring Potential Solutions. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2025; 118:e70045. [PMID: 40001298 DOI: 10.1002/arch.70045] [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: 11/20/2024] [Revised: 01/27/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025]
Abstract
Insecticides play a crucial role as the primary means of controlling agricultural pests, preventing significant damage to crops. However, the misuse of these insecticides has led to the development of resistance in insect pests against major classes of these chemicals. The emergence of resistance poses a serious threat, especially when alternative options for crop protection are limited for farmers. Addressing this challenge and developing new, effective, and sustainable pest management approaches is not merely essential but also critically important. In the absence of alternative solutions, understanding the root causes behind the development of resistance in insects becomes a critical necessity. Without this understanding, the formulation of effective approaches to combat resistance remains elusive. With insecticides playing a vital role in global food security and public health, understanding and mitigating resistance are paramount. Given the growing concern over insect resistance to insecticides, this review addresses a crucial research gap by thoroughly examining the causes, mechanisms, and potential solutions. The review examines factors driving resistance, such as evolutionary pressure and excessive pesticide use, and provides a detailed analysis of mechanisms, including detoxifying enzyme overproduction and target site mutations. Providing an analysis of potential solutions, it discusses integrated pest management, strategic insecticide rotation, and the use of new pest control technologies and biological agents. Emphasizing the urgency of a multifaceted approach, the review provides a concise roadmap for sustainable pest management, guiding future research and applications.
Collapse
Affiliation(s)
- Jiyun Liang
- College of Plant Protection, Jilin Agricultural University, Changchun, China
| | - Feng Xiao
- College of Plant Protection, Jilin Agricultural University, Changchun, China
| | - James Ojo
- Department of Crop Production, Kawara State University, Malete, Nigeria
| | - Wu Hai Chao
- College of Plant Protection, Jilin Agricultural University, Changchun, China
| | - Bilal Ahmad
- College of Plant Protection, Jilin Agricultural University, Changchun, China
| | - Aleena Alam
- College of Plant Protection, Jilin Agricultural University, Changchun, China
| | - Sohail Abbas
- College of Plant Protection, Jilin Agricultural University, Changchun, China
| | - Mogeda M Abdelhafez
- Plant Protection Research Institute, Agriculture Research Centre, Giza, Egypt
| | - Nadeemur Rahman
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Khalid Ali Khan
- Center of Bee Research and its Products and Research Centre for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
- Applied College, King Khalid University, Abha, Saudi Arabia
| | - Hamed A Ghramh
- Center of Bee Research and its Products and Research Centre for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Jamin Ali
- College of Plant Protection, Jilin Agricultural University, Changchun, China
| | - Rizhao Chen
- College of Plant Protection, Jilin Agricultural University, Changchun, China
| |
Collapse
|
143
|
Sannigrahi A, De N, Bhunia D, Bhadra J. Peptide nucleic acids: Recent advancements and future opportunities in biomedical applications. Bioorg Chem 2025; 155:108146. [PMID: 39817998 DOI: 10.1016/j.bioorg.2025.108146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/27/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025]
Abstract
Peptide nucleic acids (PNA), synthetic molecules comprising a peptide-like backbone and natural and unnatural nucleobases, have garnered significant attention for their potential applications in gene editing and other biomedical fields. The unique properties of PNA, particularly enhanced stability/specificity/affinity towards targeted DNA and RNA sequences, achieved significant attention recently for gene silencing, gene correction, antisense therapy, drug delivery, biosensing and other various diagnostic aspects. This review explores the structure, properties, and potential of PNA in transforming genetic engineering including potent biomedical challenges. In Addition, we explore future perspectives and potential limitations of PNA-based technologies, highlighting the need for further research and development to fully realize their therapeutic and biotechnological potential.
Collapse
Affiliation(s)
- Achinta Sannigrahi
- University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Nayan De
- Institute for System Biology, 401 Terry Ave N, Seattle, WA 98109, USA
| | - Debmalya Bhunia
- Cold Spring Harbor Laboratory, 1 Bungtown Rd, Cold Spring Harbor, NY 11724, USA.
| | - Jhuma Bhadra
- Department of Chemistry, Sarojini Naidu College for Women, Kolkata 700028, India.
| |
Collapse
|
144
|
Ajoolabady A, Pratico D, Mazidi M, Davies IG, Lip GYH, Seidah N, Libby P, Kroemer G, Ren J. PCSK9 in metabolism and diseases. Metabolism 2025; 163:156064. [PMID: 39547595 DOI: 10.1016/j.metabol.2024.156064] [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: 08/10/2024] [Revised: 10/02/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
PCSK9 is a serine protease that regulates plasma levels of low-density lipoprotein (LDL) and cholesterol by mediating the endolysosomal degradation of LDL receptor (LDLR) in the liver. When PCSK9 functions unchecked, it leads to increased degradation of LDLR, resulting in elevated circulatory levels of LDL and cholesterol. This dysregulation contributes to lipid and cholesterol metabolism abnormalities, foam cell formation, and the development of various diseases, including cardiovascular disease (CVD), viral infections, cancer, and sepsis. Emerging clinical and experimental evidence highlights an imperative role for PCSK9 in metabolic anomalies such as hypercholesterolemia and hyperlipidemia, as well as inflammation, and disturbances in mitochondrial homeostasis. Moreover, metabolic hormones - including insulin, glucagon, adipokines, natriuretic peptides, and sex steroids - regulate the expression and circulatory levels of PCSK9, thus influencing cardiovascular and metabolic functions. In this comprehensive review, we aim to elucidate the regulatory role of PCSK9 in lipid and cholesterol metabolism, pathophysiology of diseases such as CVD, infections, cancer, and sepsis, as well as its pharmaceutical and non-pharmaceutical targeting for therapeutic management of these conditions.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Mohsen Mazidi
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK; King's College London, Department of Twin Research & Genetic Epidemiology, South Wing St Thomas', London, UK; Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ian G Davies
- School of Sport and Exercise Sciences, Faculty of Science, Liverpool John Moores University, Copperas Hill, Liverpool L3 5AJ, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart & Chest Hospital, Liverpool, UK; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Nabil Seidah
- Laboratory of Biochemical Neuroendocrinology, Montreal Clinical Research Institute (IRCM, affiliated to the University of Montreal), Montreal, QC H2W 1R7, Canada.
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
145
|
Luo X, Germer J, Burghardt T, Grau M, Lin Y, Höhn M, Lächelt U, Wagner E. Dual pH-responsive CRISPR/Cas9 ribonucleoprotein xenopeptide complexes for genome editing. Eur J Pharm Sci 2025; 205:106983. [PMID: 39647515 DOI: 10.1016/j.ejps.2024.106983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR associated (Cas) protein has been proved as a powerful tool for the treatment of genetic diseases. The Cas9 protein, when combined with single-guide RNA (sgRNA), forms a Cas9/sgRNA ribonucleoprotein (RNP) capable of targeting and editing the genome. However, the limited availability of effective carriers has restricted the broader application of CRISPR/Cas9 RNP. In this study, we evaluated dual pH-responsive amphiphilic xenopeptides (XPs) for delivering CRISPR/Cas9 RNP. These artificial lipo-XPs contain apolar cationizable lipoamino fatty acid (LAF) and polar cationizable oligoaminoethylene acid units such as succinoyl-tetraethylenepentamine (Stp) in various ratios and U-shaped topologies. The carriers were screened for functional Cas9/sgRNA RNP delivery in four different reporter cell lines, including a Duchenne muscular dystrophy (DMD) exon skipping reporter cell model. Significantly enhanced cellular uptake into HeLa cells, effective endosomal disruption in HeLa gal8-mRuby3 cells, and potent genome editing by several Cas9/sgRNA RNP complexes was observed in four different cell lines in the 5 nM sgRNA range. Comparing Cas9/sgRNA RNP complexes with Cas9 mRNA/sgRNA polyplexes in the DMD reporter cell model demonstrated similar splice site editing and high exon skipping of the two different molecular Cas9 modalities. Based on these studies, analogues of two potent U1 LAF2-Stp and LAF4-Stp2 structures were deployed, tuning the amphiphilicity of the polar Stp group by replacement with the six oligoamino acids dmGtp, chGtp, dGtp, Htp, Stt, or GEIPA. The most potent LAF2-Stp analogues (containing dGtp, chGtp or GEIPA) demonstrated further enhanced gene editing efficiency with EC50 values of 1 nM in the DMD exon skipping reporter cell line. Notably, the EC50 of LAF2-dGtp reached 0.51 nM even upon serum incubation. Another carrier (LAF4-GEIPA2) complexing Cas9/sgRNA RNP and donor DNA, facilitated up to 43 % of homology-directed repair (HDR) in HeLa eGFPd2 cells visualized by the switch from green fluorescent protein (eGFP) to blue fluorescent protein (BFP). This study presents a delivery system tunable for Cas9 RNP complexes or Cas9 RNP/donor DNA polyplexes, offering an effective and easily applicable strategy for gene editing.
Collapse
Affiliation(s)
- Xianjin Luo
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Janin Germer
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Tobias Burghardt
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Melina Grau
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Yi Lin
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Ulrich Lächelt
- Center for Nanoscience (CeNS), LMU Munich, 80799 Munich, Germany; Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, 81377 Munich, Germany; Center for Nanoscience (CeNS), LMU Munich, 80799 Munich, Germany; CNATM - Cluster for Nucleic Acid Therapeutics Munich, Germany.
| |
Collapse
|
146
|
Chopra A, Bhuvanagiri G, Natu K, Chopra A. Role of CRISPR-Cas systems in periodontal disease pathogenesis and potential for periodontal therapy: A review. Mol Oral Microbiol 2025; 40:1-16. [PMID: 39224035 DOI: 10.1111/omi.12483] [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: 06/12/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPRs) are DNA sequences capable of editing a host genome sequence. CRISPR and its specific CRISPR-associated (Cas) protein complexes have been adapted for various applications. These include activating or inhibiting specific genetic sequences or acting as molecular scissors to cut and modify the host DNA precisely. CRISPR-Cas systems are also naturally present in many oral bacteria, where they aid in nutrition, biofilm formation, inter- and intraspecies communication (quorum sensing), horizontal gene transfer, virulence, inflammation modulation, coinfection, and immune response evasion. It even functions as an adaptive immune system, defending microbes against invading viruses and foreign genetic elements from other bacteria by targeting and degrading their DNA. Recently, CRISPR-Cas systems have been tested as molecular editing tools to manipulate specific genes linked with periodontal disease (such as periodontitis) and as novel methods of delivering antimicrobial agents to overcome antimicrobial resistance. With the rapidly increasing role of CRISPR in treating inflammatory diseases, its application in periodontal disease is also becoming popular. Therefore, this review aims to discuss the different types of CRISPR-Cas in oral microbes and their role in periodontal disease pathogenesis and precision periodontal therapy.
Collapse
Affiliation(s)
- Aditi Chopra
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Geeta Bhuvanagiri
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kshitija Natu
- School of Dentistry, University of California, Los Angeles, California, USA
| | - Avneesh Chopra
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité-University Medicine Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
147
|
Balaraman AK, Babu MA, Moglad E, Mandaliya V, Rekha MM, Gupta S, Prasad GVS, Kumari M, Chauhan AS, Ali H, Goyal K. Exosome-mediated delivery of CRISPR-Cas9: A revolutionary approach to cancer gene editing. Pathol Res Pract 2025; 266:155785. [PMID: 39708520 DOI: 10.1016/j.prp.2024.155785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Several molecular strategies based on targeted gene delivery systems have been developed in recent years; however, the CRISPR-Cas9 technology introduced a new era of targeted gene editing, precisely modifying oncogenes, tumor suppressor genes, and other regulatory genes involved in carcinogenesis. However, efficiently and safely delivering CRISPR-Cas9 to cancer cells across the cell membrane and the nucleus is still challenging. Using viral vectors and nanoparticles presents issues of immunogenicity, off-target effects, and low targeting affinity. Naturally, extracellular vesicles called exosomes have garnered the most attention as delivery vehicles in oncology-related CRISPR-Cas9 calls due to their biocompatibility, loading capacity, and inherent targeting features. The following review discusses the current progress in using exosomes to deliver CRISPR-Cas9 components, the approaches to load the CRISPR components into exosomes, and the modification of exosomes to increase stability and tumor-targeted delivery. We discuss the latest strategies in targeting recently accomplished in the exosome field, including modifying the surface of exosomes to enhance their internalization by cancer cells, as well as the measures taken to overcome the impacts of TME on delivery efficiency. Focusing on in vitro and in vivo experimentation, this review shows that exosome-mediated CRISPR-Cas9 can potentially treat cancer types, including pancreatic, lymphoma, and leukemia, for given gene targets. This paper compares exosome-mediated delivery and conventional vectors regarding safety, immune response, and targeting ability. Last but not least, we present the major drawbacks and potential development of the seemingly promising field of exosome engineering in gene editing, with references to CRISPR technologies and applications that may help make the target exosomes therapeutic in oncology.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Viralkumar Mandaliya
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Mukesh Kumari
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India.
| |
Collapse
|
148
|
Yook G, Nam J, Jo Y, Yoon H, Yang D. Metabolic engineering approaches for the biosynthesis of antibiotics. Microb Cell Fact 2025; 24:35. [PMID: 39891166 PMCID: PMC11786382 DOI: 10.1186/s12934-024-02628-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/18/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Antibiotics have been saving countless lives from deadly infectious diseases, which we now often take for granted. However, we are currently witnessing a significant rise in the emergence of multidrug-resistant (MDR) bacteria, making these infections increasingly difficult to treat in hospitals. MAIN TEXT The discovery and development of new antibiotic has slowed, largely due to reduced profitability, as antibiotics often lose effectiveness quickly as pathogenic bacteria evolve into MDR strains. To address this challenge, metabolic engineering has recently become crucial in developing efficient enzymes and cell factories capable of producing both existing antibiotics and a wide range of new derivatives and analogs. In this paper, we review recent tools and strategies in metabolic engineering and synthetic biology for antibiotic discovery and the efficient production of antibiotics, their derivatives, and analogs, along with representative examples. CONCLUSION These metabolic engineering and synthetic biology strategies offer promising potential to revitalize the discovery and development of new antibiotics, providing renewed hope in humanity's fight against MDR pathogenic bacteria.
Collapse
Affiliation(s)
- Geunsoo Yook
- Synthetic Biology and Enzyme Engineering Laboratory, Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jiwoo Nam
- Synthetic Biology and Enzyme Engineering Laboratory, Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yeonseo Jo
- Synthetic Biology and Enzyme Engineering Laboratory, Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunji Yoon
- Synthetic Biology and Enzyme Engineering Laboratory, Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Dongsoo Yang
- Synthetic Biology and Enzyme Engineering Laboratory, Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
149
|
Robertson NR, Lee S, Tafrishi A, Wheeldon I. Advances in CRISPR-enabled genome-wide screens in yeast. FEMS Yeast Res 2025; 25:foaf013. [PMID: 40113237 PMCID: PMC11995697 DOI: 10.1093/femsyr/foaf013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/12/2025] [Accepted: 03/19/2025] [Indexed: 03/22/2025] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-Cas genome-wide screens are powerful tools for unraveling genotype-phenotype relationships, enabling precise manipulation of genes to study and engineer industrially useful traits. Traditional genetic methods, such as random mutagenesis or RNA interference, often lack the specificity and scalability required for large-scale functional genomic screens. CRISPR systems overcome these limitations by offering precision gene targeting and manipulation, allowing for high-throughput investigations into gene function and interactions. Recent work has shown that CRISPR genome editing is widely adaptable to several yeast species, many of which have natural traits suited for industrial biotechnology. In this review, we discuss recent advances in yeast functional genomics, emphasizing advancements made with CRISPR tools. We discuss how the development and optimization of CRISPR genome-wide screens have enabled a host-first approach to metabolic engineering, which takes advantage of the natural traits of nonconventional yeast-fast growth rates, high stress tolerance, and novel metabolism-to create new production hosts. Lastly, we discuss future directions, including automation and biosensor-driven screens, to enhance high-throughput CRISPR-enabled yeast engineering.
Collapse
Affiliation(s)
- Nicholas R Robertson
- Bioengineering, University of California, Riverside, Riverside, CA 92521, United States
| | - Sangcheon Lee
- Chemical and Environmental Engineering, University of California, Riverside, Riverside, CA 92521, United States
| | - Aida Tafrishi
- Chemical and Environmental Engineering, University of California, Riverside, Riverside, CA 92521, United States
| | - Ian Wheeldon
- Chemical and Environmental Engineering, University of California, Riverside, Riverside, CA 92521, United States
- Center for Industrial Biotechnology, University of California, Riverside, Riverside, CA 92521, United States
| |
Collapse
|
150
|
Lee H, Rho WY, Kim YH, Chang H, Jun BH. CRISPR-Cas9 Gene Therapy: Non-Viral Delivery and Stimuli-Responsive Nanoformulations. Molecules 2025; 30:542. [PMID: 39942646 PMCID: PMC11820414 DOI: 10.3390/molecules30030542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/07/2025] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
The CRISPR-Cas9 technology, one of the groundbreaking genome editing methods for addressing genetic disorders, has emerged as a powerful, precise, and efficient tool. However, its clinical translation remains hindered by challenges in delivery efficiency and targeting specificity. This review provides a comprehensive analysis of the structural features, advantages, and potential applications of various non-viral and stimuli-responsive systems, examining recent progress to emphasize the potential to address these limitations and advance CRISPR-Cas9 therapeutics. We describe how recent reports emphasize that nonviral vectors, including lipid-based nanoparticles, extracellular vesicles, polymeric nanoparticles, gold nanoparticles, and mesoporous silica nanoparticles, can offer diverse advantages to enhance stability, cellular uptake, and biocompatibility, based on their structures and physio-chemical stability. We also summarize recent progress on stimuli-responsive nanoformulations, a type of non-viral vector, to introduce precision and control in CRISPR-Cas9 delivery. Stimuli-responsive nanoformulations are designed to respond to pH, redox states, and external triggers, facilitate controlled and targeted delivery, and minimize off-target effects. The insights in our review suggest future challenges for clinical applications of gene therapy technologies and highlight the potential of delivery systems to enhance CRISPR-Cas9's clinical efficacy, positioning them as pivotal tools for future gene-editing therapies.
Collapse
Affiliation(s)
- Hyunwoo Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (H.L.); (Y.-H.K.)
| | - Won-Yeop Rho
- School of International Engineering and Science, Jeonbuk National University, Jeonju 54896, Republic of Korea;
| | - Yoon-Hee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (H.L.); (Y.-H.K.)
| | - Hyejin Chang
- Division of Science Education, Kangwon National University, 1 Gangwondaehakgil, Chuncheon-si 24341, Republic of Korea
| | - Bong-Hyun Jun
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea; (H.L.); (Y.-H.K.)
| |
Collapse
|