1
|
Ling LA, Boukhalfa A, Kung AH, Yang VK, Chen HH. Advances in Targeted Autophagy Modulation Strategies to Treat Cancer and Associated Treatment-Induced Cardiotoxicity. Pharmaceuticals (Basel) 2025; 18:671. [PMID: 40430490 PMCID: PMC12114528 DOI: 10.3390/ph18050671] [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: 03/31/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Autophagy, an evolutionarily conserved process, plays an important role in cellular homeostasis and human diseases. Cardiovascular dysfunction, which presents during cancer treatment or in cancer-free individuals years after treatment, is a growing clinical challenge. Millions of cancer survivors and patients face an unpredictable risk of developing cardiotoxicity. Cardiotoxicity due to cancer treatment, as well as cancer progression, has been linked to autophagy dysregulation. Modulating autophagy has been further proposed as a therapeutic treatment for both cancer and cardiovascular disorders. The safe and effective use of autophagy modulation as a cardioprotective strategy during cancer treatment especially requires careful consideration and experimentation to minimize the impact on cancer treatment. We focus here on recent advances in targeted autophagy modulation strategies that utilize interdisciplinary approaches in biomedical sciences and are potentially translatable to treat cardiotoxicity and improve cancer treatment outcomes. This review highlights non-small molecule autophagy modulators to enhance targeted therapy, nanomedicine for autophagy modulation and monitoring, and in vitro models and future experiments needed to bring novel autophagy discoveries from basic research to clinical translation.
Collapse
Affiliation(s)
- Lauren A. Ling
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, #80, Boston, MA 02111, USA; (L.A.L.); (A.B.)
- School of Medicine, Tufts University, 145 Harrison Avenue, Boston, MA 02111, USA
| | - Asma Boukhalfa
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, #80, Boston, MA 02111, USA; (L.A.L.); (A.B.)
| | - Andrew H. Kung
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, #80, Boston, MA 02111, USA; (L.A.L.); (A.B.)
| | - Vicky K. Yang
- Cummings School of Veterinary Medicine, Tufts University, 200 Westboro Rd., North Grafton, MA 01536, USA;
| | - Howard H. Chen
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, #80, Boston, MA 02111, USA; (L.A.L.); (A.B.)
- School of Medicine, Tufts University, 145 Harrison Avenue, Boston, MA 02111, USA
| |
Collapse
|
2
|
Guo W, Li K, Yu H, Chang C, Zhu J, Dai K, Jiang C. The Reversible and Background-Free Hydrogel-Sensing Platform for Dual-Mode Detection of Acetone in Exhaled Breath. Anal Chem 2025; 97:4084-4091. [PMID: 39947718 DOI: 10.1021/acs.analchem.4c06189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
The acetone present in exhaled breath is a promising indicator for diagnosing human health. The fluorescent hydrogel sensor-based portable sensing platform is a highly effective tool for the on-site detection of acetone. However, existing hydrogel sensors are often limited by their irreversibility and autofluorescence. This study constructed an upconversion nanoprobe with reversibility for dual-mode detection of acetone by simply combining upconversion nanoparticles (UCNPs), hydroxylamine sulfate, and thymol blue (TB). The nanoprobe was further embedded into a hydrogel network to construct the background-free hydrogel nanosensor for the portable detection of acetone. The hydrogel nanosensor utilized long-wavelength-excited UCNPs to avoid self-luminescence interference. Hydroxylamine sulfate, as a specific recognition unit, reacted with acetone to induce the protonation of TB, resulting in an increase in absorbance at 548 nm and a decrease in luminescence at 540 nm, enabling visual colorimetric and precise luminescent detection of acetone. Moreover, the hydrogel nanosensor could be restored to its initial state through the deprotonation of TB, thereby achieving reversible detection. Additionally, 3D printing technology was utilized to construct a portable sensing platform for real-time acetone monitoring. The proposed upconversion hydrogel nanosensor in this study paves a new way for developing hydrogel sensors with high sensitivity and reversibility.
Collapse
Affiliation(s)
- Wenshuai Guo
- Anhui Province Key Laboratory of Pollutant Sensitive Materials and Environmental Remediation, Anhui Province Industrial Generic Technology Research Center for Alumics Materials, School of Physics and Electronic Information, Huaibei Normal University, Huaibei 235000, PR China
| | - Kangran Li
- Anhui Province Key Laboratory of Pollutant Sensitive Materials and Environmental Remediation, Anhui Province Industrial Generic Technology Research Center for Alumics Materials, School of Physics and Electronic Information, Huaibei Normal University, Huaibei 235000, PR China
| | - Hao Yu
- Anhui Province Key Laboratory of Pollutant Sensitive Materials and Environmental Remediation, Anhui Province Industrial Generic Technology Research Center for Alumics Materials, School of Physics and Electronic Information, Huaibei Normal University, Huaibei 235000, PR China
| | - Caidie Chang
- Anhui Province Key Laboratory of Pollutant Sensitive Materials and Environmental Remediation, Anhui Province Industrial Generic Technology Research Center for Alumics Materials, School of Physics and Electronic Information, Huaibei Normal University, Huaibei 235000, PR China
| | - Jiawei Zhu
- Anhui Province Key Laboratory of Pollutant Sensitive Materials and Environmental Remediation, Anhui Province Industrial Generic Technology Research Center for Alumics Materials, School of Physics and Electronic Information, Huaibei Normal University, Huaibei 235000, PR China
| | - Kai Dai
- Anhui Province Key Laboratory of Pollutant Sensitive Materials and Environmental Remediation, Anhui Province Industrial Generic Technology Research Center for Alumics Materials, School of Physics and Electronic Information, Huaibei Normal University, Huaibei 235000, PR China
| | - Changlong Jiang
- Key Laboratory of Photovoltaic and Energy Conservation Materials, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| |
Collapse
|
3
|
Zhang J, Tang K, Yang Y, Yang D, Fan W. Advanced Nanoprobe Strategies for Imaging Macrophage Polarization in Cancer Immunology. RESEARCH (WASHINGTON, D.C.) 2025; 8:0622. [PMID: 39990770 PMCID: PMC11842672 DOI: 10.34133/research.0622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/20/2025] [Accepted: 02/01/2025] [Indexed: 02/25/2025]
Abstract
Macrophages are ubiquitous within the human body and serve pivotal roles in immune surveillance, inflammation, and tissue homeostasis. Phenotypic plasticity is a hallmark of macrophages, allowing their polarization into distinct phenotypes M1 (pro-inflammatory, anti-tumor) and M2 (anti-inflammatory, pro-tumor) in response to local microenvironmental cues. In tumor tissues, the polarization of tumor-associated macrophages profoundly shapes the tumor microenvironment, influencing tumor progression, immune evasion, and metastasis. Therefore, the ability to image and monitor macrophage polarization is essential for comprehending tumor biology and optimizing therapeutic strategies. With the rapid advancement of nanomedicine, a diverse array of nanoprobes has been engineered to specifically target tumor-associated macrophages, offering new avenues for noninvasive in vivo imaging and real-time monitoring of macrophage dynamics within the tumor microenvironment. This perspective highlights recent advancements in macrophage-targeting nanoprobes for imaging macrophage polarization both in vitro and in vivo. It also addresses the current challenges in the field, such as enhancing probe sensitivity, specificity, and biocompatibility, while outlining the future directions for the development of next-generation nanoprobes aimed at precision oncology.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences,
Bengbu Medical University, Bengbu 233030, P.R. China
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM),
Nanjing University of Posts & Telecommunications, Nanjing 210023, P.R. China
| | - Kaiyuan Tang
- School of Fundamental Sciences,
Bengbu Medical University, Bengbu 233030, P.R. China
| | - Yongbin Yang
- Interdisciplinary Eye Research Institute (EYE-X Institute),
Bengbu Medical University, Bengbu 233030, P.R. China
| | - Dongliang Yang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences,
Nanjing Tech University (NanjingTech), Nanjing 211816, P.R. China
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials,
China Pharmaceutical University, Nanjing 211198, P.R. China
| |
Collapse
|
4
|
Akam-Baxter E, Chen HH, Boukhalfa A, Yu A, Ling LA, Kung AH, Rodriguez SB, Yuan H, Josephson L, Sosnovik DE. Decoration of Autophagy Detecting Nanoparticle with an Anionic Fluorochrome Enhances Multispectral Characterization of Autophagosome Location and Flux. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407915. [PMID: 39723734 PMCID: PMC11821426 DOI: 10.1002/smll.202407915] [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: 09/03/2024] [Revised: 11/28/2024] [Indexed: 12/28/2024]
Abstract
Autophagy is a key biological process that has proven extremely difficult to detect noninvasively. To address this, an autophagy detecting nanoparticle (ADN) was recently developed, consisting of an iron oxide nanoparticle decorated with cathepsin-cleavable arginine-rich peptides bound to the near-infrared fluorochrome Cy5.5. Activation of the probe in autophagolysosomes results in the emission of Cy5.5 fluorescence and provides a measure of autophagosome flux. However, in the early autophagosome ADN fluorescence is silent due to fluorochrome stacking. Here, we introduce to ADN a second non-cleavable fluorophore that allows the probe to be tracked through all stages of autophagy. The nature of the secondary/tracking fluorophore has a profound effect on the activation of ADN and the emission of Cy5.5 fluorescence. The lead candidate, ADN2 (featuring AZDye546 as the secondary fluorophore) has the highest activation rate and change in Cy5.5 fluorescence. Absorbance and fluorescence spectrophotometry methods show that the negatively charged AZDye546 interacts with the positively charged polyarginine motifs of the Cy5.5-polyArg activatable fluorophore, resulting in enhanced baseline quenching of the Cy5.5 signal in the nanoprobe. Flow cytometry shows that the activation of ADN2 remains specific for autophagy and is strongly modulated by classical regulators of autophagy (starvation, bafilomycin) and genetic deletion of key autophagy proteins (ATG5, ATG7). ADN2 co-localized strongly with LC3-GFP positive autophagosomes and provided readouts of in vivo probe delivery and activation in the hearts of fed/starved mice. ADN2 enhances the ability to image autophagy without genetic transfection of cells/animals and underscores the possible effects for unanticipated interactions between fluorochromes and other moieties on the surface of decorated nanoparticles.
Collapse
Affiliation(s)
- Eman Akam-Baxter
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Institute for Innovation in Imaging, Massachusetts General Hospital, Boston MA, USA
| | - Howard H. Chen
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Asma Boukhalfa
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Ada Yu
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Lauren A. Ling
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Andrew H. Kung
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Susana Bulnes Rodriguez
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hushan Yuan
- Center for Nuclear Medicine & Theranostic Sciences & PET Core, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lee Josephson
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David E. Sosnovik
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Institute for Innovation in Imaging, Massachusetts General Hospital, Boston MA, USA
| |
Collapse
|
5
|
Liu N, Zhang B, Lin N. Review on the role of autophagy in the toxicity of nanoparticles and the signaling pathways involved. Chem Biol Interact 2025; 406:111356. [PMID: 39701490 DOI: 10.1016/j.cbi.2024.111356] [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: 10/27/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
As the development of nanotechnology, the application of nanoproducts and the advancement of nanomedicine, the contact of nanoparticles (NPs) with human body is becoming increasingly prevalent. This escalation elevates the risk of NPs exposure for workers, consumers, researchers, and both aquatic and terrestrial organisms throughout the production, usage, and disposal stages. Consequently, evaluating nanotoxicity remains critically important, though standardized assessment criteria are still lacking. The diverse and complex properties of NPs further complicate the understanding of their toxicological mechanisms. Autophagy, a fundamental cellular process, exhibits dual functions-both pro-survival and pro-death. This review offers an updated perspective on the dual roles of autophagy in nanotoxicity and examines the factors influencing autophagic responses. However, no definitive framework exists for predicting NPs-induced autophagy. Beyond the conventional autophagy pathways, the review highlights specific transcription factors activated by NPs and explores metabolic reprogramming. Particular attention is given to NPs-induced selective autophagy, including mitophagy, ER-phagy, ferritinophagy, lysophagy, and lipophagy. Additionally, the review investigates autophagy's involvement in NPs-mediated biological processes such as ferroptosis, inflammation, macrophage polarization, epithelial-mesenchymal transition, tumor cell proliferation and drug resistance, as well as liver and kidney injury, neurotoxicity, and other diseases. In summary, this review presents a novel update on selective autophagy-mediated nanotoxicity and elucidates the broader interactions of autophagy in NPs-induced biological processes. Collectively, these insights offer valuable strategies for mitigating nanotoxicity through autophagy modulation and advancing the development of NPs in biomedical applications.
Collapse
Affiliation(s)
- Na Liu
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Bo Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China.
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China.
| |
Collapse
|
6
|
Zhang L, Yao X, Peng M, Ma Y, Peng Y, Wang YW. An HPQ-based near-infrared dye for the detection of ClO - and accurate imaging of cells and mouse tumor sites in situ. Org Biomol Chem 2025; 23:721-727. [PMID: 39620660 DOI: 10.1039/d4ob01599a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
A new fluorescent probe (SWJT-32) with a large Stokes shift was designed and synthesized with 2-(2-hydroxyphenyl)-4(1H)-quinazolinone (HPQ) as the skeleton and N,N-dimethylthiocarbamate as the recognition site. It can detect hypochlorite ions (ClO-) through fluorescence spectroscopy based on the excited-state intramolecular proton transfer (ESIPT) and intramolecular charge transfer (ICT) mechanism. The probe has the advantages of strong diffusion resistance, good photostability and selectivity, and high imaging contrast, and can be used for the detection of ClO-. Meanwhile, due to its low toxicity and good biocompatibility in living cells, SWJT-32 can be used for the detection of endogenous and exogenous ClO- in HeLa cells as well as for long-term in vivo imaging in tumor mice.
Collapse
Affiliation(s)
- Lu Zhang
- School of Chemistry & School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China.
| | - Xue Yao
- School of Chemistry & School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China.
| | - Min Peng
- School of Chemistry & School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China.
| | - Yu Ma
- School of Chemistry & School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China.
| | - Yu Peng
- School of Chemistry & School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China.
| | - Ya-Wen Wang
- School of Chemistry & School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China.
| |
Collapse
|
7
|
Ma M, Zhao R, Li X, Jing M, Song R, Fan J. Biological Properties of Arginine-rich Peptides and their Application in Cargo Delivery to Cancer. Curr Drug Deliv 2025; 22:387-400. [PMID: 37073158 DOI: 10.2174/1567201820666230417083350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/13/2023] [Accepted: 02/27/2023] [Indexed: 04/20/2023]
Abstract
Cell-penetrating peptides (CPPs) comprise short peptides of fewer than 30 amino acids, which are rich in arginine (Arg) or lysine (Lys). CPPs have attracted interest in the delivery of various cargos, such as drugs, nucleic acids, and other macromolecules over the last 30 years. Among all types of CPPs, arginine-rich CPPs exhibit higher transmembrane efficiency due to bidentate bonding between their guanidinium groups and negatively charged cellular components. Besides, endosome escape can be induced by arginine-rich CPPs to protect cargo from lysosome-dependent degradation. Here we summarize the function, design principles, and penetrating mechanisms of arginine-rich CPPs, and outline their biomedical applications in drug delivery and biosensing in tumors.
Collapse
Affiliation(s)
- Minghai Ma
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ruizhao Zhao
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
- Clinical Medical School, Xi'an Medical University, Xi'an, 710061, China
| | - Xing Li
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Minxuan Jing
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Rundong Song
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jinhai Fan
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
8
|
Draper I, Huang W, Pande S, Zou A, Calamaras TD, Choe RH, Correia-Branco A, Mei AL, Chen HH, Littel HR, Gunasekaran M, Wells NM, Bruels CC, Daugherty AL, Wolf MJ, Kang PB, Yang VK, Slonim DK, Wallingford MC, Blanton RM. The splicing factor hnRNPL demonstrates conserved myocardial regulation across species and is altered in heart failure. FEBS Lett 2024; 598:2670-2682. [PMID: 39300280 PMCID: PMC11560511 DOI: 10.1002/1873-3468.15020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/22/2024]
Abstract
Heart failure (HF) is highly prevalent. Mechanisms underlying HF remain incompletely understood. Splicing factors (SF), which control pre-mRNA alternative splicing, regulate cardiac structure and function. This study investigated regulation of the splicing factor heterogeneous nuclear ribonucleoprotein-L (hnRNPL) in the failing heart. hnRNPL protein increased in left ventricular tissue from mice with transaortic constriction-induced HF and from HF patients. In left ventricular tissue, hnRNPL was detected predominantly in nuclei. Knockdown of the hnRNPL homolog Smooth in Drosophila induced cardiomyopathy. Computational analysis of predicted mouse and human hnRNPL binding sites suggested hnRNPL-mediated alternative splicing of tropomyosin, which was confirmed in C2C12 myoblasts. These findings identify hnRNPL as a sensor of cardiac dysfunction and suggest that disturbances of hnRNPL affect alternative splicing in HF.
Collapse
Affiliation(s)
- Isabelle Draper
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Wanting Huang
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Suchita Pande
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Aaron Zou
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Timothy D Calamaras
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Richard H Choe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | | | - Ariel L Mei
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Howard H Chen
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Hannah R Littel
- Greg Marzolf Jr. Muscular Dystrophy Center, Department of Neurology, and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Mekala Gunasekaran
- Greg Marzolf Jr. Muscular Dystrophy Center, Department of Neurology, and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Natalya M Wells
- Greg Marzolf Jr. Muscular Dystrophy Center, Department of Neurology, and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Christine C Bruels
- Greg Marzolf Jr. Muscular Dystrophy Center, Department of Neurology, and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Audrey L Daugherty
- Greg Marzolf Jr. Muscular Dystrophy Center, Department of Neurology, and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Matthew J Wolf
- Division of Cardiology, University of Virginia, Charlottesville, VA, USA
| | - Peter B Kang
- Greg Marzolf Jr. Muscular Dystrophy Center, Department of Neurology, and Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Vicky K Yang
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, MA, USA
| | | | - Robert M Blanton
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
9
|
Beesabathuni NS, Kenaston MW, Gangaraju R, Adia NAB, Peddamallu V, Shah PS. Let's talk about flux: the rising potential of autophagy rate measurements in disease. Autophagy 2024; 20:2574-2580. [PMID: 38984617 PMCID: PMC11572197 DOI: 10.1080/15548627.2024.2371708] [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: 07/05/2023] [Revised: 06/10/2024] [Accepted: 06/19/2024] [Indexed: 07/11/2024] Open
Abstract
Macroautophagy/autophagy is increasingly implicated in a variety of diseases, making it an attractive therapeutic target. However, many aspects of autophagy are not fully understood and its impact on many diseases remains debatable and context-specific. The lack of systematic and dynamic measurements in these cases is a key reason for this ambiguity. In recent years, Loos et al. 2014 and Beesabathuni et al. 2022 developed methods to quantitatively measure autophagy holistically. In this commentary, we pose some of the unresolved biological questions regarding autophagy and consider how quantitative measurements may address them. While the applications are ever-expanding, we provide specific use cases in cancer, virus infection, and mechanistic screening. We address how the rate measurements themselves are central to developing cancer therapies and present ways in which these tools can be leveraged to dissect the complexities of virus-autophagy interactions. Screening methods can be combined with rate measurements to mechanistically decipher the labyrinth of autophagy regulation in cancer and virus infection. Taken together, these approaches have the potential to illuminate the underlying mechanisms of various diseases.Abbreviation MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; R1: rate of autophagosome formation; R2: rate of autophagosome-lysosome fusion; R3: rate of autolysosome turnover.
Collapse
Affiliation(s)
| | - Matthew W. Kenaston
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA, USA
| | - Ritika Gangaraju
- Department of Chemical Engineering, University of California, Davis, CA, USA
| | - Neil Alvin B. Adia
- Department of Chemical Engineering, University of California, Davis, CA, USA
| | - Vardhan Peddamallu
- Department of Chemical Engineering, University of California, Davis, CA, USA
| | - Priya S. Shah
- Department of Chemical Engineering, University of California, Davis, CA, USA
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA, USA
| |
Collapse
|
10
|
Mei L, Liao K, Chen H, Zhang Y, Zhang Z, Li Q, Li M. Application of Nanomaterials and Related Drug Delivery Systems in Autophagy. Molecules 2024; 29:3513. [PMID: 39124918 PMCID: PMC11313712 DOI: 10.3390/molecules29153513] [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: 05/15/2024] [Revised: 07/12/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Autophagy, a lysosomal self-degradation pathway, plays a critical role in cellular homeostasis by degrading endogenous damaged organelles and protein aggregates into recyclable biological molecules. Additionally, it detoxifies extracellular toxic substances, including drugs and toxic materials, thereby preserving the stability of the intracellular environment. The swift progression of nanotechnology has led to an increased focus on understanding the relationship between nanomaterials and autophagy. The effects of various nanomaterials and nano drug delivery systems on autophagy and their biological functions have been preliminarily assessed, revealing that modulation of intracellular autophagy levels by these agents represents a novel cellular response mechanism. Notably, autophagy regulation based on nanomaterials or nano drug delivery systems for a range of diseases is currently the subject of extensive research. Given the close association between autophagy levels and tumors, the regulation of autophagy has emerged as a highly active area of research in the development of innovative tumor therapies. This review synthesizes the current understanding of the application of nanomaterials or nano drug delivery systems on autophagy and their potential biological functions, suggesting a new avenue for nanomaterial-based autophagy regulation.
Collapse
Affiliation(s)
- Ling Mei
- Engineering Research Center for Pharmaceuticals and Equipment of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; (L.M.)
| | - Kai Liao
- Engineering Research Center for Pharmaceuticals and Equipment of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; (L.M.)
| | - Haiyan Chen
- Engineering Research Center for Pharmaceuticals and Equipment of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; (L.M.)
| | - Yifan Zhang
- Engineering Research Center for Pharmaceuticals and Equipment of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; (L.M.)
| | - Zihan Zhang
- Engineering Research Center for Pharmaceuticals and Equipment of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; (L.M.)
| | - Qiangwei Li
- Engineering Research Center for Pharmaceuticals and Equipment of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; (L.M.)
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Luo H, Tian L, Zhang Y, Wu Y, Li B, Liu J. Recent advances in molecular and nanoparticle probes for fluorescent bioanalysis. NANO RESEARCH 2024; 17:6443-6474. [DOI: 10.1007/s12274-024-6659-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 01/06/2025]
|
12
|
Xu Y, Chen J, Zhang Y, Zhang P. Recent Progress in Peptide-Based Molecular Probes for Disease Bioimaging. Biomacromolecules 2024; 25:2222-2242. [PMID: 38437161 DOI: 10.1021/acs.biomac.3c01413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Recent strides in molecular pathology have unveiled distinctive alterations at the molecular level throughout the onset and progression of diseases. Enhancing the in vivo visualization of these biomarkers is crucial for advancing disease classification, staging, and treatment strategies. Peptide-based molecular probes (PMPs) have emerged as versatile tools due to their exceptional ability to discern these molecular changes with unparalleled specificity and precision. In this Perspective, we first summarize the methodologies for crafting innovative functional peptides, emphasizing recent advancements in both peptide library technologies and computer-assisted peptide design approaches. Furthermore, we offer an overview of the latest advances in PMPs within the realm of biological imaging, showcasing their varied applications in diagnostic and therapeutic modalities. We also briefly address current challenges and potential future directions in this dynamic field.
Collapse
Affiliation(s)
- Ying Xu
- School of Biomedical Engineering and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Junfan Chen
- School of Biomedical Engineering and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Yuan Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Pengcheng Zhang
- School of Biomedical Engineering and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
13
|
Huang J, Zhang D, Zu Y, Zhang L. Procalcitonin Detection Using Immunomagnetic Beads-Mediated Surface-Enhanced Raman Spectroscopy. BIOSENSORS 2024; 14:164. [PMID: 38667157 PMCID: PMC11048292 DOI: 10.3390/bios14040164] [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: 02/26/2024] [Revised: 03/19/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024]
Abstract
The early detection of procalcitonin (PCT) is crucial for diagnosing bacterial infections due to its high sensitivity and specificity. While colloidal gold colorimetric and immune-chemiluminescence methods are commonly employed in clinical detection, the former lacks sensitivity, and the latter faces challenges with a brief luminescence process and an elevated background. Here, we introduce a novel approach for the quantitative analysis of PCT using surface-enhanced Raman spectroscopy (SERS), leveraging the enhanced properties of metal nanoparticles. Simultaneously, we employed a magnetic nanoparticle coating and surface biofunctionalization modification to immobilize PCT-trapping antibodies, creating the required immune substrates. The resulting magnetic nanoparticles and antibody complexes, acting as carriers and recognition units, exhibited superparamagnetism and the specific recognition of biomarkers. Then, this complex efficiently underwent magnetic separation with an applied magnetic field, streamlining the cumbersome steps of traditional ELISA and significantly reducing the detection time. In conclusion, the exploration of immunomagnetic bead detection technology based on surface-enhanced Raman spectroscopy holds crucial practical significance for the sensitive detection of PCT.
Collapse
Affiliation(s)
- Jiayue Huang
- Joint Centre of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China;
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative In-novation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Dagan Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yan Zu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Lexiang Zhang
- Joint Centre of Translational Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China;
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| |
Collapse
|
14
|
Xie C, Peng Y, Zhang Z, Luo K, Yang Q, Tan L, Zhou L. Tumor Microenvironment Activatable Nanoprodrug System for In Situ Fluorescence Imaging and Therapy of Liver Cancer. Anal Chem 2024; 96:5006-5013. [PMID: 38484040 DOI: 10.1021/acs.analchem.4c00317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
The development of new imaging and treatment nanoprodrug systems is highly demanded for diagnosis and therapy of liver cancer, a severe disease characterized by a high recurrence rate. Currently, available small molecule drugs are not possible for cancer diagnosis because of the fast diffusion of imaging agents and low efficacy in treatment due to poor water solubility and significant toxic side effects. In this study, we report the development of a tumor microenvironment activatable nanoprodrug system for the diagnosis and treatment of liver cancer. This nanoprodrug system can accumulate in the tumor site and be selectively activated by an excess of hydrogen peroxide (H2O2) in the tumor microenvironment, releasing near-infrared solid-state organic fluorescent probe (HPQCY-1) and phenylboronic acid-modified camptothecin (CPT) prodrug. Both HPQCY-1 and CPT prodrugs can be further activated in tumor sites for achieving more precise in situ near-infrared (NIR) fluorescence imaging and treatment while reducing the toxic effects of drugs on normal tissues. Additionally, the incorporation of hydrophilic multivalent chitosan as a carrier effectively improved the water solubility of the system. This research thus provides a practical new approach for the diagnosis and treatment of liver cancer.
Collapse
Affiliation(s)
- Can Xie
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan 410004, China
| | - Yongbo Peng
- The Key Laboratory of Biochemistry and Mo-lecular Pharmacology, Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Zhen Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Kun Luo
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan 410004, China
| | - Qiaomei Yang
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan 410004, China
| | - Libin Tan
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan 410004, China
| | - Liyi Zhou
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan 410004, China
| |
Collapse
|
15
|
Yan K, Hu Z, Yu P, He Z, Chen Y, Chen J, Sun H, Wang S, Zhang F. Ultra-photostable small-molecule dyes facilitate near-infrared biophotonics. Nat Commun 2024; 15:2593. [PMID: 38519530 PMCID: PMC10960032 DOI: 10.1038/s41467-024-46853-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 03/12/2024] [Indexed: 03/25/2024] Open
Abstract
Long-wavelength, near-infrared small-molecule dyes are attractive in biophotonics. Conventionally, they rely on expanded aromatic structures for redshift, which comes at the cost of application performance such as photostability, cell permeability, and functionality. Here, we report a ground-state antiaromatic strategy and showcase the concise synthesis of 14 cationic aminofluorene dyes with mini structures (molecular weights: 299-504 Da) and distinct spectra covering 700-1600 nm. Aminofluorene dyes are cell-permeable and achieve rapid renal clearance via a simple 44 Da carboxylation. This accelerates optical diagnostics of renal injury by 50 min compared to existing macromolecular approaches. We develop a compact molecular sensing platform for in vivo intracellular sensing, and demonstrate the versatile applications of these dyes in multispectral fluorescence and optoacoustic imaging. We find that aromaticity reversal upon electronic excitation, as indicated by magnetic descriptors, not only reduces the energy bandgap but also induces strong vibronic coupling, resulting in ultrafast excited-state dynamics and unparalleled photostability. These results support the argument for ground-state antiaromaticity as a useful design rule of dye development, enabling performances essential for modern biophotonics.
Collapse
Affiliation(s)
- Kui Yan
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, PR China
| | - Zhubin Hu
- State Key Laboratory of Precision Spectroscopy, School of Physics and Electronic Science, East China Normal University, Shanghai, PR China
| | - Peng Yu
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, PR China
| | - Zuyang He
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, PR China
| | - Ying Chen
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, PR China
| | - Jiajian Chen
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, PR China
| | - Haitao Sun
- State Key Laboratory of Precision Spectroscopy, School of Physics and Electronic Science, East China Normal University, Shanghai, PR China.
| | - Shangfeng Wang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, PR China.
| | - Fan Zhang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem, Fudan University, Shanghai, PR China.
| |
Collapse
|
16
|
Tang J, Yoon N, Dadson K, Sung HK, Lei Y, Dang TQ, Chung WY, Ahmed S, Abdul-Sater AA, Wu J, Li RK, Jonkman J, McKee T, Grant J, Peterson JD, Sweeney G. Impaired autophagy flux contributes to enhanced ischemia reperfusion injury in the diabetic heart. AUTOPHAGY REPORTS 2024; 3:2330327. [PMID: 40395534 PMCID: PMC11864616 DOI: 10.1080/27694127.2024.2330327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 02/07/2024] [Accepted: 03/05/2024] [Indexed: 05/22/2025]
Abstract
Myocardial ischemia/reperfusion (I/R) injury is exacerbated in diabetic individuals and animal models. We tested whether autophagy is an important cellular determinant of cell death. First, we utilized a cellular model of hypoxia reoxygenation (H/R) in H9c2 cells cultured in low or high glucose (HG) and tested cell death using flow cytometry to detect Annexin-V and propidium iodide, imaging cell viability ReadyProbe and lactate dehydrogenase release. We observed that cell death induced by H/R was enhanced by HG. Kinetic analysis of caspase-3 activity using a fluorescence reporter probe, stable expression of the VC3AI biosensor and western blotting indicated that H/R induced activation of caspase-3 was enhanced by HG. Temporal autophagy flux analysis using DapRed and DalGreen probes indicated an initial increase in response to H/R that was reduced upon prolonged (24h) R. HG suppressed this induction of autophagy. This was verified using LC3 HiBiT reporter assay, tandem-fluorescent LC3, and western blotting. Lysosomal cathepsin activity was also elevated at 6h and suppressed at 24h R. Autophagy-deficient cells were generated via CRISPR-mediated knockout of atg7 and the effect of combined HG and H/R treatment on caspase activation and cell death was elevated in comparison with wild type cells. We then performed coronary artery ligation surgery to induce ischemia, followed by reperfusion, in wild-type or streptozotocin (STZ)-induced hyperglycemic mice. Non-invasive 3-dimensional imaging using fluorescence molecular tomography combined with computerized tomography was employed to monitor spatio-temporal activation of cardiac autophagy and apoptosis. Upon systemic injection of a near infra-red cathepsin activatable probe we found that hyperglycemic mice had lower activity in the infarct region after I/R versus wild type. In parallel, we observed a higher extent of I/R-induced apoptosis, detected with an annexin-V probe, in hyperglycemic mice. Collectively, these results revealed that impaired autophagic flux in the presence of high glucose levels exacerbates I/R injury.Abbreviation: satg7, autophagy-related 7; FMT, fluorescence molecular tomography; HG, high glucose; H/R, hypoxia/reoxygenation; I/R, ischemia/reperfusion; LC3, MAP1LC3; N, normoxia; NG, normal glucose; NIR, near-infrared; p62, SQSTM1; STZ, streptozotocin.
Collapse
Affiliation(s)
- Jialing Tang
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Nanyoung Yoon
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Keith Dadson
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Hye Kyoung Sung
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Yubin Lei
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Thanh Q. Dang
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Wing Yan Chung
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Saher Ahmed
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Ali A. Abdul-Sater
- School of Kinesiology and Health Science, York University, Toronto, Canada
| | - Jun Wu
- Division of Cardiovascular Surgery and Toronto General Research Institute, University Health Network, Toronto, Canada
| | - Ren-Ke Li
- Division of Cardiovascular Surgery and Toronto General Research Institute, University Health Network, Toronto, Canada
| | - James Jonkman
- Advanced Optical Microscopy Facility (AOMF), University Health Network, Toronto, Canada
| | - Trevor McKee
- Spatio-temporal Targeting and Amplification of Radiation Response (STTARR), Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Justin Grant
- Spatio-temporal Targeting and Amplification of Radiation Response (STTARR), Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario
| | | | - Gary Sweeney
- Department of Biology, York University, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Sung HK, Tang J, Jahng JWS, Song E, Chan YK, Lone AH, Peterson J, Abdul‐Sater A, Sweeney G. Ischemia-induced cardiac dysfunction is exacerbated in adiponectin-knockout mice due to impaired autophagy flux. Clin Transl Sci 2024; 17:e13758. [PMID: 38515365 PMCID: PMC10958170 DOI: 10.1111/cts.13758] [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: 09/01/2023] [Revised: 01/23/2024] [Accepted: 02/14/2024] [Indexed: 03/23/2024] Open
Abstract
Strategies to enhance autophagy flux have been suggested to improve outcomes in cardiac ischemic models. We explored the role of adiponectin in mediating cardiac autophagy under ischemic conditions induced by permanent coronary artery ligation. We studied the molecular mechanisms underlying adiponectin's cardio-protective effects in adiponectin knockout (Ad-KO) compared with wild-type (WT) mice subjected to ischemia by coronary artery ligation and H9c2 cardiomyocyte cell line exposed to hypoxia. Systemic infusion of a cathepsin-B activatable near-infrared probe as a biomarker for autophagy and detection via noninvasive three-dimensional fluorescence molecular tomography combined with computerized tomography to quantitate temporal changes, indicated increased activity in the myocardium of WT mice after myocardial infarction which was attenuated in Ad-KO. Seven days of ischemia increased myocardial adiponectin accumulation and elevated ULK1/AMPK phosphorylation and autophagy assessed by Western blotting for LC3 and p62, an outcome not observed in Ad-KO mice. Cell death, assessed by TUNEL analysis and the ratio of Bcl-2:Bax, plus cardiac dysfunction, measured using echocardiography with strain analysis, were exacerbated in Ad-KO mice. Using cellular models, we observed that adiponectin stimulated autophagy flux in isolated primary adult cardiomyocytes and increased basal and hypoxia-induced autophagy in H9c2 cells. Real-time temporal analysis of caspase-3/7 activation and caspase-3 Western blot indicated that adiponectin suppressed activation by hypoxia. Hypoxia-induced mitochondrial reactive oxygen species production and cell death were also attenuated by adiponectin. Importantly, the ability of adiponectin to reduce caspase-3/7 activation and cell death was not observed in autophagy-deficient cells generated by CRISPR-mediated deletion of Atg7. Collectively, our data indicate that adiponectin acts in an autophagy-dependent manner to attenuate cardiomyocyte caspase-3/7 activation and cell death in response to hypoxia in vitro and ischemia in mice.
Collapse
Affiliation(s)
| | - Jialing Tang
- Department of BiologyYork UniversityTorontoOntarioCanada
| | | | - Erfei Song
- Department of BiologyYork UniversityTorontoOntarioCanada
| | - Yee Kwan Chan
- Department of BiologyYork UniversityTorontoOntarioCanada
| | | | | | - Ali Abdul‐Sater
- School of Kinesiology and Health ScienceYork UniversityTorontoOntarioCanada
| | - Gary Sweeney
- Department of BiologyYork UniversityTorontoOntarioCanada
| |
Collapse
|
18
|
Pan L, Peng H, Lee B, Zhao J, Shen X, Yan X, Hua Y, Kim J, Kim D, Lin M, Zhang S, Li X, Yi X, Yao F, Qin Z, Du J, Chi Y, Nam JM, Hyeon T, Liu J. Cascade Catalytic Nanoparticles Selectively Alkalize Cancerous Lysosomes to Suppress Cancer Progression and Metastasis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2305394. [PMID: 37643367 DOI: 10.1002/adma.202305394] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/16/2023] [Indexed: 08/31/2023]
Abstract
Lysosomes are critical in modulating the progression and metastasis for various cancers. There is currently an unmet need for lysosomal alkalizers that can selectively and safely alter the pH and inhibit the function of cancer lysosomes. Here an effective, selective, and safe lysosomal alkalizer is reported that can inhibit autophagy and suppress tumors in mice. The lysosomal alkalizer consists of an iron oxide core that generates hydroxyl radicals (•OH) in the presence of excessive H+ and hydrogen peroxide inside cancer lysosomes and cerium oxide satellites that capture and convert •OH into hydroxide ions. Alkalized lysosomes, which display impaired enzyme activity and autophagy, lead to cancer cell apoptosis. It is shown that the alkalizer effectively inhibits both local and systemic tumor growth and metastasis in mice. This work demonstrates that the intrinsic properties of nanoparticles can be harnessed to build effective lysosomal alkalizers that are both selective and safe.
Collapse
Affiliation(s)
- Limin Pan
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Haibao Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Bowon Lee
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jiaxu Zhao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Xiulian Shen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ximei Yan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yipeng Hua
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Jeonghyun Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dokyoon Kim
- Department of Bionano Engineering, Hanyang University, Ansan, 15588, Republic of Korea
| | - Mouhong Lin
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Shengjian Zhang
- Department of Radiology, Cancer Hospital/Institute and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaohui Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Xueying Yi
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Feibai Yao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhiyong Qin
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yudan Chi
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Jwa-Min Nam
- Department of Chemistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Taeghwan Hyeon
- Center for Nanoparticle Research, Institute for Basic Science (IBS), Seoul, 08826, Republic of Korea
- School of Chemical and Biological Engineering, and Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jianan Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
19
|
Liu J, Li L, Zhang R, Xu ZP. The adjacent effect between Gd(III) and Cu(II) in layered double hydroxide nanoparticles synergistically enhances T1-weighted magnetic resonance imaging contrast. NANOSCALE HORIZONS 2023; 8:279-290. [PMID: 36606452 DOI: 10.1039/d2nh00478j] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Magnetic resonance imaging (MRI) is one key technology in modern diagnostic medicine. However, the development of high-relaxivity contrast agents with favorable properties for imaging applications remains a challenging task. In this work, dual Gd(III) and Cu(II) doped-layered double hydroxide (GdCu-LDH) nanoparticles show significantly higher longitudinal relaxivity compared with sole-metal-based LDH (Gd-LDH and Cu-LDH) nanoparticles. This relaxation enhancement in GdCu-LDH is also much greater than the simple addition of the relaxivity rate of the two paramagnetic ions in Gd-LDH and Cu-LDH, presumably attributed to synergistic T1 shortening between adjacent Gd(III) and Cu(II) in the LDH host layers (adjacent effect). Moreover, our GdCu-LDH nanoparticles exhibit a pH-ultrasensitive property in MRI performance and show much clearer MR imaging for tumor tissues in mice than Gd-LDH and Cu-LDH at the equivalent doses. Thus, these novel Gd/Cu-co-doped LDH nanoparticles provide higher potential for accurate cancer diagnosis in clinic application. To the best of our knowledge, this is the first report that two paramagnetic metal ions in one nanoparticle synergistically improve the T1-MRI contrast.
Collapse
Affiliation(s)
- Jianping Liu
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD 4072, Australia.
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD 4072, Australia.
| | - Run Zhang
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD 4072, Australia.
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD 4072, Australia.
- Institute of Biomedical Health Technology and Engineering and Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, P. R. China, 518107
| |
Collapse
|
20
|
Loos B, du Toit A, Hofmeyr JHS. Non-invasive monitoring of autophagy. Nat Biomed Eng 2022; 6:1015-1016. [PMID: 36127452 DOI: 10.1038/s41551-022-00943-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa.
| | - Andre du Toit
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | | |
Collapse
|