1
|
Hu C, Yang Z, Shi X, Xue Y, Huang L, Tang C, Wang F. In vivo monitoring of endogenous hydrogen sulfide and evaluation of natural protectants in liver injury mice using a highly selective bioluminescent probe. Biosens Bioelectron 2025; 278:117343. [PMID: 40058276 DOI: 10.1016/j.bios.2025.117343] [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: 01/13/2025] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/30/2025]
Abstract
Hydrogen sulfide (H2S) is an essential endogenous gasotransmitter that can regulate a wide range of physiological processes. However, overproduction of H2S is toxic to humans and causes liver injury, cardiovascular diseases, central nervous system-related disease, diabetes, even cancer. Hence, designing efficient imaging probes for real-time monitoring of the alterations in endogenous H2S is a viable tactic for accurate diagnosis of these diseases. In this work, a bioluminescence (BL) probe, namely Luc-H2S, has been developed to achieve H2S detection in vitro and in vivo. This sensing probe enables a selective BL turn-on response to H2S, and showcases excellent sensitivity with a low detection limit (LOD = 0.337 μM). Furthermore, Luc-H2S has been successfully applied in BL imaging of endogenous H2S in cells, tumor-bearing mice and drug-induced liver injury mice. More importantly, Luc-H2S is utilized to accurately evaluate the protective effects of natural products against alcohol-induced liver injury through monitoring of the H2S fluctuations. We envision that Luc-H2S holds promise as a powerful imaging tool for diagnosis of H2S-mediated diseases and evaluation of drug therapy.
Collapse
Affiliation(s)
- Chong Hu
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Zeping Yang
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Xiaorui Shi
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Yaru Xue
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Liyu Huang
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Chu Tang
- Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China.
| | - Fu Wang
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China; Institute of Medical Engineering, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
2
|
Zheng Y, Ma X, Zhou S, Lei W, Luo X, Zhou L, Xu K, Zhong W. Alkaline Phosphatase and ATG4B Sequentially Activated Fluorescent Probe for Cancer Cell-Specific Live Imaging of Autophagy. Anal Chem 2025; 97:8370-8377. [PMID: 40197017 DOI: 10.1021/acs.analchem.4c06950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Tracking autophagy in cancer cells is crucial for enhancing cancer therapies. Existing methods are often inefficient and cannot distinguish cancer from normal cells during autophagy. Herein, a sequentially activated peptide probe, NBD-1p-Dabcyl, was developed for achieving cancer cell-specific imaging of autophagy. The probe self-assembled and fluoresced brightly upon sequential processing by alkaline phosphatase (ALP) and autophagy-related protease (ATG4B), where NBD-1p-Dabcyl was dephosphorylated by ALP to give NBD-1-Dabcyl, which was then processed by ATG4B into nanofibers emitting strong fluorescence. Notably, the bright fluorescence of NBD was observed in cancer cells MDA-MB-231 and HeLa, while normal cells NIH3T3 exhibited weaker fluorescence, allowing differentiation between cancer and normal cells using a rapamycin (Rap)-induced autophagy cell model. The enhanced fluorescence in cancer cells was attributed to the higher activities of intracellular ALP and ATG4B. Next, NBD-1p-Dabcyl was used to assess the inhibition efficiency of an autophagy inhibitor NSC 185058 in MDA-MB-231 cells, where a strong correlation between fluorescence intensity and inhibitor concentration suggested that NBD-1p-Dabcyl could predict the activity of autophagy inhibitors. Finally, animal experiments revealed that NBD-1p-Dabcyl effectively facilitated in situ fluorescence imaging of autophagy in tumor tissues. The design of this sequentially activated peptide probe offers a practical approach for monitoring autophagy in cancer cells, enabling high-throughput screening of autophagy inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Yaxin Zheng
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Xinyue Ma
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Shuyao Zhou
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Wenwen Lei
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Xuan Luo
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Zhou
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 210009, China
| | - Keming Xu
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 210009, China
| | - Wenying Zhong
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
3
|
Jia H, Wang C, Fu Y, Wang Y, Zhang X, Tang Y, Ding J, He K, Wang J, Shen Y. Visualization of mitochondrial molecular dynamics during mitophagy process by label-free surface-enhanced Raman scattering spectroscopy. Anal Chim Acta 2025; 1345:343748. [PMID: 40015786 DOI: 10.1016/j.aca.2025.343748] [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: 10/30/2024] [Revised: 01/18/2025] [Accepted: 01/29/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Mitophagy is a selective way to eliminate dysfunctional mitochondria and recycle their constituents, which plays an important role in regulating and maintaining intracellular homeostasis. Real-time monitoring mitophagy process is of great importance for cellular physiological and pathological processes related to mitochondria. Howbeit, most of the current methods only focus on single-parameter detection of mitochondrial microenvironmental changes such as pH, viscosity and polarity. The mitochondrial molecular responses under mitophagy are not clear. Therefore, developing a new and simple method for molecular profiling is of great importance for accurately and comprehensively visualizing mitophagy. RESULTS In this work, Au NPs-based mitochondria-targeting nanoprobe was developed and the nanoprobe-based label-free surface enhanced Raman spectroscopy (SERS) method was proposed to track starvation induced mitophagy process at molecular level. The nanoprobe displayed good SERS performance and low cytotoxicity. Based on the developed strategy, the molecular response within mitochondria under mitophagy was validated. Meanwhile, the protein denaturation, conformational change, lipid degradation and DNA fragmentation within mitochondria under mitophagy were revealed for the first time, which provides molecular evidence for mitophagy. The changes in reactive oxygen species level and mitochondrial membrane potential further confirmed the damage of mitochondria. Moreover, the developed label-free SERS strategy was used to detect mitophagy in drug (cisplatin)-induced liver injury (DILI) cell model, and obvious mitophagy in DILI cells was observed. SIGNIFICANCE The molecular biochemical signature dynamic changes within mitochondria during mitophagy process were revealed by SERS for the first time. Moreover, compared with the current research, our study can provide new insights into mitophagy and mitophagy-involved diseases at molecular level. This study will provide new insights into the molecular mechanism of mitophagy and offer a simple and effective method for mitochondrial molecular event monitoring in mitophagy-involved cellular processes.
Collapse
Affiliation(s)
- Hailan Jia
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Chi Wang
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yan Fu
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yalin Wang
- The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xiaoyu Zhang
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yuezhou Tang
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jiahao Ding
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Kun He
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jing Wang
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, People's Republic of China.
| | - Yanting Shen
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang, People's Republic of China.
| |
Collapse
|
4
|
Zhang Y, Gu W, Chen W, Zhu J, Fan L, Zhang L, Zhao L, Miao Q. A Dual-Targeted Molecule for Disease-Activatable Proteolysis Targeting Chimeras and Targeted Radionuclide Therapy of Cancer. J Am Chem Soc 2025; 147:7897-7907. [PMID: 39989465 DOI: 10.1021/jacs.4c18398] [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: 02/25/2025]
Abstract
Proteolysis targeting chimeras (PROTACs) represent a cutting-edge approach for targeted protein degradation in cancer therapy, yet they face challenges such as poor pharmacokinetics and specificity issues, leading to undesirable off-target effects and limited antitumor potency. To address these issues, we introduce dual-targeted unimolecular theranostic probes (e.g., radioactive 177Lu-P-A and its cold counterpart natLu-P-A) for disease-activatable PROTACs in combination with targeted radionuclide therapy (TRT) against prostate cancer with high specificity and effectiveness. The probes achieve a cathepsin B (CTSB)-activatable pro-PROTAC moiety for precise degradation of bromodomain-containing protein 4 (BRD4) and a prostate-specific membrane antigen (PSMA)-targeted 177Lu-based TRT. Owing to the favorable pharmacokinetics and PSMA-mediated excellent targeting efficiency, the probe possesses high tumor imaging specificity and accumulation capacity of therapeutic units for highly effective PROTACs and TRT. In contrast, the free PROTACs unit (e.g., ARV-771) shows no observable therapeutic effect due to its poor targeting ability. Importantly, the BRD4 proteolysis by PROTAC activation can downregulate radiosensitivity-associated RAD51AP1 expression, synergistically enhancing the TRT effect and promoting apoptosis after combined therapy compared to individual treatment regimes. Additionally, the probe demonstrates high renal clearance, underscoring its biosafety for potential clinical translation. This study presents a potential approach for precise PROTACs combined with TRT for effective tumor therapy.
Collapse
Affiliation(s)
- Yuan Zhang
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei 230026, China
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Wei Gu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Wan Chen
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jieli Zhu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Longfei Fan
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Liwen Zhang
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Liangyou Zhao
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Qingqing Miao
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei 230026, China
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
5
|
Sun C, Liu S, Lau JW, Yang H, Chen Y, Xing B. Enzyme-Activated Orthogonal Proteolysis Chimeras for Tumor Microenvironment-Responsive Immunomodulation. Angew Chem Int Ed Engl 2025:e202423057. [PMID: 39932237 DOI: 10.1002/anie.202423057] [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: 11/26/2024] [Indexed: 02/20/2025]
Abstract
Precise modulation of dynamic and complex tumor microenvironment (TME) to disrupt tumorigenesis and reshape intratumoral immune infiltration has emerged as promising approaches for enhanced cancer therapy. Among recent innovations, proteolysis-targeting chimeras (PROTACs) represent a burgeoning chemical knockdown technology capable of degrading oncogenic protein homeostasis and inducing dynamic alternations within carcinoma settings, offering potential for antitumor manipulation. However, achieving selectivity in PROTACs that respond to disease environmental stimulation and precisely perturb on-target proteins remains challenging. The multi-step synthesis and limited permeability, attributed to high-molecular-weight and heterobifunctional structures, further hinder their in vivo efficacy. Herein, we present a unique TME-responsive enzyme-activated clickable PROTACs, which features a short peptide-tagged pomalidomide derivative to undergo tumor-specific cleavage by cathepsin protease to induce orthogonal crosslinking of the exposed cysteine with 2-cyanobenzothiazole-labeled epigenetic protein-ligand JQ1, facilitating in situ degrader formation within tumor regions only. Systematic protein profiling and proteomic analysis revealed that such TME-specific clickable-PROTACs not only selectively eliminate epigenetic proteins without tedious pre-synthesis to bridge disparate small-molecule bi-warhead fragments, but also demonstrated superior tumor penetration compared to conventional high-molecular-weight PROTACs. Importantly, these clickable-PROTACs efficiently downregulated immune checkpoint programmed death-ligand 1 (PD-L1) both in vitro and in vivo, remodeling TME for enhanced therapeutics, especially in anti-tumoral immunomodulation.
Collapse
Affiliation(s)
- Caixia Sun
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Songhan Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Jun Wei Lau
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | - Hanyu Yang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Yun Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Bengang Xing
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Kowloon, Hong Kong SAR, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| |
Collapse
|
6
|
Liu Y, Yao Y, Sha J, Liang G, Sun X. Dual-Locked Enzyme-Activatable Fluorescence Probes for Precise Bioimaging. ACS Biomater Sci Eng 2025; 11:730-741. [PMID: 39841057 DOI: 10.1021/acsbiomaterials.4c01858] [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: 01/23/2025]
Abstract
Real-time visualization of endogenous enzymes not only helps reveal the underlying biological principles but also provides pathological information for cancer/disease diagnosis and even treatment guidance. To this end, enzyme-activatable fluorescence probes are frequently fabricated that turn their fluorescence signals "on" exclusively at the enzyme-rich region, thus enabling noninvasive and real-time imaging of enzymes of interest at the molecular level with superior sensitivity and selectivity. However, in a complex biological context, commonly used single enzyme-activatable (i.e., single-locked) probes may suffer from "false positive" signals at healthy tissues and be insufficient to accurately indicate the occurrence of certain diseases. Therefore, dual-locked fluorescence probes have been promoted to address these issues. Using dual enzymes (or an enzyme with another stimulus) as "keys", they permit simultaneous detection of distinct biomarkers, offering significantly enhanced imaging precision toward certain biological events. Considering that recent reviews on these probes remain scarce, we thus provide this review. We summarize the recent progress, particularly highlighting the breakthroughs in the last three years, and discuss the challenges in this field.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Yuchen Yao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Junhui Sha
- School of Life Science and Technology, Southeast University, Nanjing 211189, China
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
- Handan Norman Technology Company, Limited, Guantao 057750, China
| | - Xianbao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
7
|
Swartchick C, Dirak M, Wenger LCF, Tapia Hernandez R, Lee MC, Chan J. Activity-Based Bioluminescent Logic-Gate Probe Reveals Crosstalk Between the Inflammatory Tumor Microenvironment and ALDH1A1 in Cancer Cells. JACS AU 2025; 5:320-331. [PMID: 39886570 PMCID: PMC11775703 DOI: 10.1021/jacsau.4c01001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 02/01/2025]
Abstract
Cancer cells with high expression of aldehyde dehydrogenase 1A1 (ALDH1A1) are more resistant to chemotherapy, contribute to tumor progression, and are associated with poor clinical outcomes. ALDH1A1 plays a critical role in protecting cells from reactive aldehydes and, in the case of stem cells, regulates their differentiation through the retinoic acid signaling pathway. Despite the importance of this enzyme, methods to study ALDH1A1 high-expressing cancer cells in vivo remain limited. In this work, we developed AlDeLuc, the first logic-gated bioluminescence probe designed to selectively evaluate ALDH1A1 activity in tumor cells. The probe is sequentially activated by acidic intracellular compartments (i.e., endosomes) and ALDH1A1, ensuring precise detection of ALDH1A1 high-expressing cells and minimizing off-target detection of non-ALDH1A1 cells. Beyond demonstrating efficacy in multiple cancer cell lines and a murine model of breast cancer, we employed AlDeLuc to investigate how the population of ALDH1A1 high-expressing cells is influenced by the inflammatory status of a tumor in the context of a high-fat diet. These findings establish a molecular link between obesity, inflammation, and tumor progression.
Collapse
Affiliation(s)
- Chelsea
B. Swartchick
- Department of Chemistry,
Beckman Institute for Advanced Science and Technology, and Cancer
Center at Illinois, University of Illinois
Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Musa Dirak
- Department of Chemistry,
Beckman Institute for Advanced Science and Technology, and Cancer
Center at Illinois, University of Illinois
Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Lily C. F. Wenger
- Department of Chemistry,
Beckman Institute for Advanced Science and Technology, and Cancer
Center at Illinois, University of Illinois
Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Rodrigo Tapia Hernandez
- Department of Chemistry,
Beckman Institute for Advanced Science and Technology, and Cancer
Center at Illinois, University of Illinois
Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Michael C. Lee
- Department of Chemistry,
Beckman Institute for Advanced Science and Technology, and Cancer
Center at Illinois, University of Illinois
Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Jefferson Chan
- Department of Chemistry,
Beckman Institute for Advanced Science and Technology, and Cancer
Center at Illinois, University of Illinois
Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
8
|
Dai SY, Xiao Z, Shen F, Lim I, Rao J. Light-Controlled Intracellular Synthesis of Poly(luciferin) Polymers Induces Cell Paraptosis. J Am Chem Soc 2025; 147:2037-2048. [PMID: 39757486 DOI: 10.1021/jacs.4c15644] [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: 01/07/2025]
Abstract
Accumulation of misfolded proteins challenges cellular proteostasis and is implicated in aging and chronic disorders. Cancer cells, moreover, face an elevated level of basal proteotoxic stress; hence, exacerbating endoplasmic reticulum (ER) stress has been shown to induce programmed cell death while enhancing anticancer immunogenicity. We hypothesize that hydrophobic abiotic macromolecules can trigger a similar stress response. Most polymers and nanoparticles, however, are sequestered in endo/lysosomes after endocytosis, which prevents their interaction with the proteostasis machinery. We adopted an in situ polymerization approach to synthesize polymers in cells with cell-permeable monomers. Specifically, we developed a biocompatible polycondensation between l-cysteine and 2-cyanobenzothiazole (CBT) with photochemical control to form insoluble poly(luciferin) aggregates. We identified that in situ polymerization activates the BiP-PERK-CHOP pathway of the unfolded protein response and that the unresolved ER stress initiates a form of regulated cell death consistent with paraptosis. In addition, the dying cells emit damage-associated molecular patterns (DAMPs), indicating an immunogenic cell death that could potentiate antitumor immunity. Our results show that in situ polymerization mimics misfolded protein aggregates to induce proteotoxic stress and cancer cell death, offering a novel therapeutic strategy to exploit cancer vulnerability.
Collapse
Affiliation(s)
- Sheng-Yao Dai
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Zhen Xiao
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Fangfang Shen
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Irene Lim
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Jianghong Rao
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
9
|
Yuan Z, Jiang Q, Liang G. Inspired by nature: Bioluminescent systems for bioimaging applications. Talanta 2025; 281:126821. [PMID: 39255622 DOI: 10.1016/j.talanta.2024.126821] [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: 06/04/2024] [Revised: 09/01/2024] [Accepted: 09/05/2024] [Indexed: 09/12/2024]
Abstract
Bioluminescence is a natural process where biological organisms produce light through chemical reactions. These reactions predominantly occur between small-molecule substrates and luciferase within bioluminescent organisms. Bioluminescence imaging (BLI) has shown significant potential in biomedical research owing to its non-invasive, real-time observation and quantification. In this review, we introduced the chemical mechanism of bioluminescent systems and categorized several strategies that successfully addressed the native limitations, including improvements on the chemical structures of luciferase-luciferin bioluminescence system and bioluminescence resonance energy transfer (BRET) methods. In addition, we also reviewed and summarized recent advances in bioimaging applications. We hope that this review can provide effective guidance for the development and application of bioluminescent systems in the field of bioimaging.
Collapse
Affiliation(s)
- Zihan Yuan
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Qiaochu Jiang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China; Handan Norman Technology Co., Ltd., Guantao, 057750, China.
| |
Collapse
|
10
|
Wang R, Zhou L, Yang Y, Zhao F, Sun X, Liu X, Zou Z, Liang G. Spatially Quantitative Imaging of Enzyme Activity in a Living Cell. J Am Chem Soc 2024; 146:34870-34877. [PMID: 39655641 DOI: 10.1021/jacs.4c14190] [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: 12/19/2024]
Abstract
Enzyme activity plays a key role in cell heterogeneity. Its spatially quantitative imaging in a living cell not only directly displays but also helps people to understand cell heterogeneity. Current methods are hard to achieve due to the short intracellular retention or lack of internal reference of the imaging probes. Herein, we rationally designed a self-referenced Raman probe Val-Cit-Cys(StBu)-Pra-Gly-CBT (Yne-CBT) which takes an intracellular cathepsin B (CTSB)-initiated CBT-Cys click reaction to yield a long-retained cyclic dimer in cell. In the meantime, Raman signal changes of its two chemical bonds (C≡C and C≡N) after the reaction are used for self-referencing and quantitative Raman imaging of CTSB activity. In vitro experiments demonstrated that, with shell-isolated nanoparticle-enhanced Raman spectroscopy technique, 20 μM Yne-CBT was able to quantitatively detect CTSB activity with a limit of detection of 61.4 U L-1. Under a homemade microfluidic channel, Yne-CBT was successfully applied for spatially quantitative imaging CTSB activity in a living cell. Our strategy provides people with a facile method to directly and quantitatively display cell heterogeneity.
Collapse
Affiliation(s)
- Rui Wang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Lei Zhou
- School of Science, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Yueyan Yang
- School of Science, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Furong Zhao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xianbao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xiaoyang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Zhen Zou
- Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research, Ministry of Education, Institute of Interdisciplinary Studies, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, Hunan 410081, China
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| |
Collapse
|
11
|
Hong Y, Yang Y, Wang C, Huang Y, Shen W, Shen Z, Lun Z, Zhang J, Wang C, Yuan Y. Luciferase-Loaded Calcium Phosphate Nanoparticles for Persistent Bioluminescence Imaging of Orthotopic Breast Tumors. Anal Chem 2024; 96:14320-14325. [PMID: 39208257 DOI: 10.1021/acs.analchem.4c02289] [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: 09/04/2024]
Abstract
Bioluminescence imaging (BLI) is an important noninvasive optical imaging technique that has been widely used to monitor many biological processes due to its high sensitivity, resolution, and signal-to-noise ratio. However, the BLI technique based on the firefly luciferin-luciferase system is limited by the expression of exogenous luciferase and the short half-life of firefly luciferin, which pose challenges for long-term tracking in vivo. To solve the problems, here we rationally designed an intelligent strategy for persistent BLI in tumors by combining luciferase-loaded calcium phosphate nanoparticles (Luc@CaP NPs) to provide luciferase and the probe Cys(SEt)-Lys-CBT (CKCBT) to slowly produce the luciferase substrate amino luciferin (Am-luciferin). Luc@CaP NPs constructed with CaP as a carrier could enable luciferase activity to be maintained in vivo for at least 12 h. And compared to the conventional substrate luciferin, CKCBT apparently prolonged the BL time by up to 2 h through GSH-induced intracellular self-assembly and subsequent protease degradation-induced release of Am-luciferin. We anticipate that this strategy could be applied for clinical translation in more disease diagnosis and treatment in the near future.
Collapse
Affiliation(s)
- Yajian Hong
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230031, China
- Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yanyun Yang
- Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Chenchen Wang
- Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yifan Huang
- Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Weicheng Shen
- Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Zhiqiang Shen
- Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Zhiyou Lun
- Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jia Zhang
- Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Congxiao Wang
- Department of the Interventional Medical Center, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Yue Yuan
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230031, China
- Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
12
|
Liu F, Zhou T, Zhang S, Li Y, Chen Y, Miao Z, Wang X, Yang G, Li Q, Zhang L, Liu Y. Cathepsin B: The dawn of tumor therapy. Eur J Med Chem 2024; 269:116329. [PMID: 38508117 DOI: 10.1016/j.ejmech.2024.116329] [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: 11/10/2023] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
Cathepsin B (CTSB) is a key lysosomal protease that plays a crucial role in the development of cancer. This article elucidates the relationship between CTSB and cancer from the perspectives of its structure, function, and role in tumor growth, migration, invasion, metastasis, angiogenesis and autophagy. Further, we summarized the research progress of cancer treatment related drugs targeting CTSB, as well as the potential and advantages of Traditional Chinese medicine in treating tumors by regulating the expression of CTSB.
Collapse
Affiliation(s)
- Fuxian Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Ting Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China; Experimental & Training Teaching Centers, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shangzu Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yangyang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yan Chen
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhiming Miao
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Xin Wang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Gengqiang Yang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Qiyang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China
| | - Liying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China; College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China.
| | - Yongqi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou, China; College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China; Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou, China.
| |
Collapse
|