0
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Nanoparticles systemically biodistribute to regenerating skeletal muscle in DMD

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Skeletal muscle disease severity can often progress asymmetrically across muscle groups and heterogeneously within tissues. An example is Duchenne Muscular Dystrophy (DMD) in which lack of dystrophin results in devastating skeletal muscle wasting in some muscles whereas others are spared or undergo hypertrophy. An efficient, non-invasive approach to identify sites of asymmetry and degenerative lesions could enable better patient monitoring and therapeutic targeting of disease. In this study, we utilized a versatile intravenously injectable mesoporous silica nanoparticle (MSNP) based nanocarrier system to explore mechanisms of biodistribution in skeletal muscle of mdx mouse models of DMD including wildtype, dystrophic, and severely dystrophic mice. Moreover, MSNPs could be imaged in live mice and whole muscle tissues enabling investigation of how biodistribution is altered by different types of muscle pathology such as inflammation or fibrosis. We found MSNPs were tenfold more likely to aggregate within select mdx muscles relative to wild type, such as gastrocnemius and quadriceps. This was accompanied by decreased biodistribution in off-target organs. We found the greatest factor affecting preferential delivery was the regenerative state of the dystrophic skeletal muscle with the highest MSNP abundance coinciding with the regions showing the highest level of embryonic myosin staining and intramuscular macrophage uptake. To demonstrate, muscle regeneration regulated MSNP distribution, we experimentally induced regeneration using barium chloride which resulted in a threefold increase of intravenously injected MSNPs to sites of regeneration 7 days after injury. These discoveries provide the first evidence that nanoparticles have selective biodistribution to skeletal muscle in DMD to areas of active regeneration and that nanoparticles could enable diagnostic and selective drug delivery in DMD skeletal muscle.

          Supplementary Information

          The online version contains supplementary material available at 10.1186/s12951-023-01994-0.

          Related collections

          Most cited references68

          • Record: found
          • Abstract: found
          • Article: not found

          Mesoporous silica nanoparticles: synthesis, biocompatibility and drug delivery.

          In the past decade, mesoporous silica nanoparticles (MSNs) have attracted more and more attention for their potential biomedical applications. With their tailored mesoporous structure and high surface area, MSNs as drug delivery systems (DDSs) show significant advantages over traditional drug nanocarriers. In this review, we overview the recent progress in the synthesis of MSNs for drug delivery applications. First, we provide an overview of synthesis strategies for fabricating ordered MSNs and hollow/rattle-type MSNs. Then, the in vitro and in vivo biocompatibility and biotranslocation of MSNs are discussed in relation to their chemophysical properties including particle size, surface properties, shape, and structure. The review also highlights the significant achievements in drug delivery using mesoporous silica nanoparticles and their multifunctional counterparts as drug carriers. In particular, the biological barriers for nano-based targeted cancer therapy and MSN-based targeting strategies are discussed. We conclude with our personal perspectives on the directions in which future work in this field might be focused. Copyright © 2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Nanoparticle-liver interactions: Cellular uptake and hepatobiliary elimination.

            30-99% of administered nanoparticles will accumulate and sequester in the liver after administration into the body. This results in reduced delivery to the targeted diseased tissue and potentially leads to increased toxicity at the hepatic cellular level. This review article focuses on the inter- and intra-cellular interaction between nanoparticles and hepatic cells, the elimination mechanism of nanoparticles through the hepatobiliary system, and current strategies to manipulate liver sequestration. The ability to solve the "nanoparticle-liver" interaction is critical to the clinical translation of nanotechnology for diagnosing and treating cancer, diabetes, cardiovascular disorders, and other diseases.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Diverse Applications of Nanomedicine

              The design and use of materials in the nanoscale size range for addressing medical and health-related issues continues to receive increasing interest. Research in nanomedicine spans a multitude of areas, including drug delivery, vaccine development, antibacterial, diagnosis and imaging tools, wearable devices, implants, high-throughput screening platforms, etc. using biological, nonbiological, biomimetic, or hybrid materials. Many of these developments are starting to be translated into viable clinical products. Here, we provide an overview of recent developments in nanomedicine and highlight the current challenges and upcoming opportunities for the field and translation to the clinic.
                Bookmark

                Author and article information

                Contributors
                MSpencer@mednet.ucla.edu
                mengh@nanoctr.cn
                apyle@mednet.ucla.edu
                Journal
                J Nanobiotechnology
                J Nanobiotechnology
                Journal of Nanobiotechnology
                BioMed Central (London )
                1477-3155
                29 August 2023
                29 August 2023
                2023
                : 21
                : 303
                Affiliations
                [1 ]GRID grid.19006.3e, ISNI 0000 0000 9632 6718, Department of Medicine, David Geffen School of Medicine at UCLA, ; Los Angeles, CA USA
                [2 ]GRID grid.509979.b, ISNI 0000 0004 7666 6191, California Nanosystems Institute at UCLA, ; Los Angeles, CA USA
                [3 ]GRID grid.19006.3e, ISNI 0000 0000 9632 6718, Department of Microbiology, Immunology and Medical Genetics, David Geffen School of Medicine at UCLA, ; Los Angeles, CA USA
                [4 ]GRID grid.475520.1, Eli and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, ; Los Angeles, CA USA
                [5 ]GRID grid.19006.3e, ISNI 0000 0000 9632 6718, Department of Neurology, David Geffen School of Medicine at UCLA, ; Los Angeles, CA USA
                [6 ]GRID grid.266093.8, ISNI 0000 0001 0668 7243, Department of Physiology and Biophysics, , University of California Irvine, ; Irvine, CA USA
                [7 ]GRID grid.9227.e, ISNI 0000000119573309, Present Address: Zhejiang Cancer Hospital, , Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, ; Hangzhou, 310022 Zhejiang China
                [8 ]Present Address: MyoGene Bio, San Diego, CA USA
                [9 ]GRID grid.419265.d, ISNI 0000 0004 1806 6075, Present Address: CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, , National Center for Nanoscience and Technology, ; Beijing, China
                Article
                1994
                10.1186/s12951-023-01994-0
                10463982
                37641124
                5aaf8968-b698-443c-8efd-e0d64f5e7fa9
                © BioMed Central Ltd., part of Springer Nature 2023

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

                History
                : 29 October 2022
                : 9 July 2023
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100000900, California Institute for Regenerative Medicine;
                Categories
                Research
                Custom metadata
                © BioMed Central Ltd., part of Springer Nature 2023

                Biotechnology
                Biotechnology

                Comments

                Comment on this article