1
|
Lim EY, Kim GD, Kim HJ, Eom JE, Song HJ, Shin DU, Kim YI, Kim HJ, Lee SY, Shin HS. Cirsium japonicum leaf extract attenuated lipopolysaccharide-induced acute respiratory distress syndrome in mice via suppression of the NLRP3 and HIF1α pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156601. [PMID: 40064116 DOI: 10.1016/j.phymed.2025.156601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/20/2024] [Accepted: 03/01/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a severe inflammatory disorder characterized by acute respiratory failure, alveolar barrier dysfunction, edema, and dysregulated alveolar macrophage-mediated pulmonary inflammation. Despite advancements in treatment strategies, the mortality rate in patients with ARDS remains high, ranging from 40-60 %. Current approaches are limited to supportive care, necessitating the exploration of effective therapeutic options such as suppressing broad inflammatory responses. Although Cirsium japonicum leaves possess anti-inflammatory properties, their specific effects on ARDS have not yet been investigated. METHODS The anti-inflammatory activity of Cirsium japonicum extract (CJE) was investigated in a lipopolysaccharide (LPS)-induced ARDS model. RESULTS CJE significantly attenuated LPS-induced lung injury, including reduced alveolar wall thickness, inflammatory cell infiltration, proteinaceous debris, and hyaline membranes. Moreover, CJE repressed infiltration of inflammatory cells and pro-inflammatory gene expression in bronchoalveolar lavage fluid. Concordantly, CJE mitigated alveolar macrophage activation, which consequently reduced neutrophil chemoattractic infiltration. Additionally, CJE suppressed NLRP3 and HIF1α expression in the lungs of the ARDS mouse. Similarly, LPS-induced NLRP3 and HIF1α pathway-associated inflammatory and glycolytic gene expressions significantly diminished by CJE in murine alveolar macrophage cell line, MH-S cells, and bone marrow-derived macrophages. CONCLUSION CJE suppressed multiple inflammatory responses through the regulation of NLRP3 and HIF1α signaling-related gene expression in macrophages of LPS-induced ARDS mice. These results suggest that CJE has therapeutic potential for treating patients with ARDS via macrophage regulation.
Collapse
Affiliation(s)
- Eun Yeong Lim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Gun-Dong Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Ha-Jung Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Ji-Eun Eom
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Hyeon-Ji Song
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea; Department of Food Science and Technology, Jeonbuk National University, Jeonju 54896, South Korea
| | - Dong-Uk Shin
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Young In Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Hyun-Jin Kim
- Department of Food Science and Technology, Gyeongsang National University, Jinju 52828, South Korea
| | - So-Young Lee
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea; Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, South Korea
| | - Hee Soon Shin
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea; Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, South Korea.
| |
Collapse
|
2
|
Akgul Obeidin SV, Senol MS, Dogru Koseoglu Z, Bayramoglu F, Disli S, Yigitbasi T, Emekli N. Antibiotic-derived approaches in cancer therapy: effectiveness of ikarugamycin in hexokinase-2 inhibition, tissue factor modulation, and metabolic regulation in breast cancer. Anticancer Drugs 2025; 36:328-337. [PMID: 39879102 DOI: 10.1097/cad.0000000000001689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
We aimed to explore the role of ikarugamycin (IKA) in breast cancer, its connection with hexokinase-2 (HK-2) repression, and tissue factor (TF). This study sought to extend the role of HK-2 as a TF activator in a comprehensive analysis of these interactions from the enzyme, gene, and protein levels. The investigation was performed with MDA-MB-231 and MCF-7 breast cancer lines. The oxidative stress index (OSI), lactate production, and HK activity were assessed using colorimetric assays. Western blot and quantitative PCR analyses were performed to determine HK-2 and TF expressions. Prothrombin time Tests additionally assessed the effect of IKA therapy on TF activation. Three over four significantly downregulated genes were identified after a specific analysis of the IKA's effect on HK-2 and TF in breast cancer cell lines. In the IKA treatment group, lactate production was markedly reduced ( P < 0.05) and hexokinase activity was found to be reduced in all groups ( P < 0.05, <0.01). Paclitaxel cytotoxicity independently causes lower OSI in all IKA-treated groups as compared to controls even though OSI is elevated in IKA groups compared to control. Molecular analysis results demonstrated significantly downregulated HK-2 and TF expressions at the protein level ( P < 0.05, P < 0.01). Partial thromboplastin time results also showed that IKA-treated cells had longer TF activation duration. A potential indirect association of HK-2 inhibition and TF regulation in breast cancer cells is put forward in this study by presenting IKA's bioactivation of breast cancer in all gene, protein, and enzyme levels.
Collapse
Affiliation(s)
| | - Masite Sehadet Senol
- Department of Biochemistry, Institute of Health Science
- Department of Biochemistry, Faculty of Medicine
| | - Zeynep Dogru Koseoglu
- Department of Biochemistry, Institute of Health Science
- Istanbul Medipol University Genetic Disorders Assessment Center (MEDIGEN), Istanbul Medipol University, Istanbul, Türkiye
| | - Feyza Bayramoglu
- Department of Biochemistry, Institute of Health Science
- Department of Biochemistry, Faculty of Medicine
| | - Sevgi Disli
- Department of Biochemistry, Institute of Health Science
- Department of Biochemistry, Faculty of Medicine
| | - Turkan Yigitbasi
- Department of Biochemistry, Institute of Health Science
- Department of Biochemistry, Faculty of Medicine
- Istanbul Medipol University Genetic Disorders Assessment Center (MEDIGEN), Istanbul Medipol University, Istanbul, Türkiye
| | - Neslin Emekli
- Department of Biochemistry, Institute of Health Science
- Department of Biochemistry, Faculty of Medicine
| |
Collapse
|
3
|
Liu Y, Hu P, Cheng H, Xu F, Ye Y. The impact of glycolysis on ischemic stroke: from molecular mechanisms to clinical applications. Front Neurol 2025; 16:1514394. [PMID: 39926015 PMCID: PMC11802445 DOI: 10.3389/fneur.2025.1514394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/10/2025] [Indexed: 02/11/2025] Open
Abstract
Ischemic stroke (IS), a leading cause of disability and mortality worldwide, remains a significant challenge due to its complex pathogenesis. Glycolysis, a central metabolic pathway, plays a critical role in bridging the gap between metabolic dysfunction and neurological impairment. During ischemic conditions, glycolysis replaces oxidative phosphorylation as the primary energy source for brain tissue. However, in the ischemia-reperfusion state, neuronal cells show a particular reliance on aerobic glycolysis. Immune cells, such as monocytes, also contribute to atheromatous plaque formation and thrombi through increased aerobic glycolysis. Given glycolysis's involvement in various pathological stages of IS, it offers the potential for improved diagnosis, treatment, and prevention. This review comprehensively explores the role of glycolysis in different phases of IS, addresses existing controversies, and discusses its diagnostic and therapeutic applications. By elucidating the intricate relationship between glycolysis and IS, this review aims to provide novel insights for future research and clinical advancements.
Collapse
Affiliation(s)
- Yingquan Liu
- The First Clinical College of Anhui University of Chinese Medicine, Hefei, China
| | - Peijia Hu
- The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hongliang Cheng
- The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Fangyuan Xu
- The First Clinical College of Anhui University of Chinese Medicine, Hefei, China
| | - Yu Ye
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
4
|
Liu Z, Zheng X, Li N, Wang Z. Baicalein suppresses inflammation and attenuates acute lung injury by inhibiting glycolysis via HIF‑1α signaling. Mol Med Rep 2025; 31:18. [PMID: 39513601 PMCID: PMC11564906 DOI: 10.3892/mmr.2024.13383] [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: 06/09/2024] [Accepted: 09/18/2024] [Indexed: 11/15/2024] Open
Abstract
Baicalein, a flavonoid monomer compound isolated from the dried root of the traditional Chinese herb Scutellaria baicalensis, has several pharmacological activities, such as anti‑inflammatory, anti‑angiogenic, antitumor, antimicrobial and antiviral properties. Acute lung injury (ALI) is characterized by injury of the alveolar epithelium and capillary endothelium, which results in decreased lung volume, decreased lung compliance, ventilation/perfusion mismatch, intrapulmonary edema, alveolar edema and even acute hypoxemic respiratory failure. The present study aimed to investigate the effects of baicalein on lung injury and inflammation. Bioinformatics analysis using network pharmacology predicted that the hypoxia inducible factor‑1α (HIF‑1α) and glycolysis signaling pathways were involved in the mechanism underlying the therapeutic effects of baicalein. Further in vitro and in vivo experiments, such as immunohistochemistry, immunofluorescence and PCR, verified that baicalein could inhibit HIF‑1α signaling, thus suppressing glycolysis, and improving inflammatory responses and ALI. Taken together, the results of the present study suggested that the anti‑inflammatory effects of baicalein on treating ALI were associated with its ability to suppress glycolysis via the HIF‑1α signaling pathway.
Collapse
Affiliation(s)
- Zhongyou Liu
- Department of Respiratory Diseases, Zhumadian Hospital of Traditional Chinese Medicine, Zhumadian, Henan 463000, P.R. China
| | - Xiaona Zheng
- Department of Respiratory Diseases, Zhumadian Hospital of Traditional Chinese Medicine, Zhumadian, Henan 463000, P.R. China
| | - Ning Li
- Department of Scientific Research, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Zongyao Wang
- Department of Respiratory Diseases, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan 450007, P.R. China
| |
Collapse
|
5
|
Anaya-Sanchez A, Berry SB, Espich S, Zilinskas A, Tran PM, Agudelo C, Samani H, Darwin KH, Portnoy DA, Stanley SA. Methylglyoxal is an antibacterial effector produced by macrophages during infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.03.621721. [PMID: 39554200 PMCID: PMC11566019 DOI: 10.1101/2024.11.03.621721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Infected macrophages transition into aerobic glycolysis, a metabolic program crucial for control of bacterial infection. However, antimicrobial mechanisms supported by aerobic glycolysis are unclear. Methylglyoxal is a highly toxic aldehyde that modifies proteins and DNA and is produced as a side-product of glycolysis. Here we show that despite the toxicity of this aldehyde, infected macrophages generate high levels of methylglyoxal during aerobic glycolysis while downregulating the detoxification system. We use targeted mutations in mice to modulate methylglyoxal generation and show that reducing methylglyoxal production by the host promotes survival of Listeria monocytogenes and Mycobacterium tuberculosis , whereas increasing methylglyoxal levels improves control of bacterial infection. Furthermore, we show that bacteria that are unable to detoxify methylglyoxal are avirulent and experience up to 1000-fold greater genomic mutation frequency during infection. Taken together, these results suggest that methylglyoxal is an antimicrobial innate immune effector that defends the host against bacterial pathogens.
Collapse
|
6
|
Peng M, Zou R, Yao S, Meng X, Wu W, Zeng F, Chen Z, Yuan S, Zhao F, Liu W. High-intensity interval training and medium-intensity continuous training may affect cognitive function through regulation of intestinal microbial composition and its metabolite LPS by the gut-brain axis. Life Sci 2024; 352:122871. [PMID: 38936602 DOI: 10.1016/j.lfs.2024.122871] [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: 03/01/2024] [Revised: 06/16/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024]
Abstract
AIMS The gut-brain axis is the communication mechanism between the gut and the central nervous system, and the intestinal flora and lipopolysaccharide (LPS) play a crucial role in this mechanism. Exercise regulates the gut microbiota composition and metabolite production (i.e., LPS). We aimed to investigate the effects of high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) on cognitive function in C57BL/6 J mice through gut-brain axis regulation of gut microbiota composition and LPS displacement. MAIN METHODS C57BL/6 J male mice were randomly divided into sedentary, HIIT, and MICT groups. After 12 weeks of exercise intervention, the cognitive function of the brain and mRNA levels of related inflammatory factors were measured. RNA sequencing, Golgi staining, intestinal microbial 16 s rDNA sequencing, and ELISA were performed. KEY FINDINGS HIIT and MICT affect brain cognitive function by regulating the gut microbiota composition and its metabolite, LPS, through the gut microbiota-gut-brain axis. HIIT is suspected to have a risk: it can induce "intestinal leakage" by regulating intestinal permeability-related microbiota, resulting in excessive LPS in the blood and brain and activating M1 microglia in the brain, leading to reduced dendritic spine density and affecting cognitive function. SIGNIFICANCE This study revealed a potential link between changes in the gut microbiota and cognitive function. It highlighted the possible risk of HIIT in reducing dendritic spine density and affecting cognitive function.
Collapse
Affiliation(s)
- Mei Peng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Ruihan Zou
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Sisi Yao
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Xiangyuan Meng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Weijia Wu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Fanqi Zeng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Zeyu Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Shunling Yuan
- Yangtze University College of Arts and Sciences, Jingzhou 434020, China
| | - Fei Zhao
- The First Affiliated Hospital of Hunan Normal University, Changsha 410002, China
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China; Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
7
|
Liu B, Zhou H, Tan L, Siu KTH, Guan XY. Exploring treatment options in cancer: Tumor treatment strategies. Signal Transduct Target Ther 2024; 9:175. [PMID: 39013849 PMCID: PMC11252281 DOI: 10.1038/s41392-024-01856-7] [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: 01/23/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 07/18/2024] Open
Abstract
Traditional therapeutic approaches such as chemotherapy and radiation therapy have burdened cancer patients with onerous physical and psychological challenges. Encouragingly, the landscape of tumor treatment has undergone a comprehensive and remarkable transformation. Emerging as fervently pursued modalities are small molecule targeted agents, antibody-drug conjugates (ADCs), cell-based therapies, and gene therapy. These cutting-edge treatment modalities not only afford personalized and precise tumor targeting, but also provide patients with enhanced therapeutic comfort and the potential to impede disease progression. Nonetheless, it is acknowledged that these therapeutic strategies still harbour untapped potential for further advancement. Gaining a comprehensive understanding of the merits and limitations of these treatment modalities holds the promise of offering novel perspectives for clinical practice and foundational research endeavours. In this review, we discussed the different treatment modalities, including small molecule targeted drugs, peptide drugs, antibody drugs, cell therapy, and gene therapy. It will provide a detailed explanation of each method, addressing their status of development, clinical challenges, and potential solutions. The aim is to assist clinicians and researchers in gaining a deeper understanding of these diverse treatment options, enabling them to carry out effective treatment and advance their research more efficiently.
Collapse
Affiliation(s)
- Beilei Liu
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Hongyu Zhou
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Licheng Tan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Kin To Hugo Siu
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China.
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, China.
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China.
| |
Collapse
|
8
|
Huang W, Fu G, Wang Y, Chen C, Luo Y, Yan Q, Liu Y, Mao C. Immunometabolic reprogramming of macrophages with inhalable CRISPR/Cas9 nanotherapeutics for acute lung injury intervention. Acta Biomater 2024; 181:308-316. [PMID: 38570107 DOI: 10.1016/j.actbio.2024.03.031] [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: 02/29/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/05/2024]
Abstract
Acute lung injury (ALI) represents a critical respiratory condition typified by rapid-onset lung inflammation, contributing to elevated morbidity and mortality rates. Central to ALI pathogenesis lies macrophage dysfunction, characterized by an overabundance of pro-inflammatory cytokines and a shift in metabolic activity towards glycolysis. This study emphasizes the crucial function of glucose metabolism in immune cell function under inflammatory conditions and identifies hexokinase 2 (HK2) as a key regulator of macrophage metabolism and inflammation. Given the limitations of HK2 inhibitors, we propose the CRISPR/Cas9 system for precise HK2 downregulation. We developed an aerosolized core-shell liposomal nanoplatform (CSNs) complexed with CaP for efficient drug loading, targeting lung macrophages. Various CSNs were synthesized to encapsulate an mRNA based CRISPR/Cas9 system (mCas9/gHK2), and their gene editing efficiency and HK2 knockout were examined at both gene and protein levels in vitro and in vivo. The CSN-mCas9/gHK2 treatment demonstrated a significant reduction in glycolysis and inflammation in macrophages. In an LPS-induced ALI mouse model, inhaled CSN-mCas9/gHK2 mitigated the proinflammatory tumor microenvironment and reprogrammed glucose metabolism in the lung, suggesting a promising strategy for ALI prevention and treatment. This study highlights the potential of combining CRISPR/Cas9 gene editing with inhalation delivery systems for effective, localized pulmonary disease treatment, underscoring the importance of targeted gene modulation and metabolic reprogramming in managing ALI. STATEMENT OF SIGNIFICANCE: This study investigates an inhalable CRISPR/Cas9 gene editing system targeting pulmonary macrophages, with the aim of modulating glucose metabolism to alleviate Acute Lung Injury (ALI). The research highlights the role of immune cell metabolism in inflammation, as evidenced by changes in macrophage glucose metabolism and a notable reduction in pulmonary edema and inflammation. Additionally, observed alterations in macrophage polarization and cytokine levels in bronchoalveolar lavage fluid suggest potential therapeutic implications. These findings not only offer insights into possible ALI treatments but also contribute to the understanding of immune cell metabolism in inflammatory diseases, which could be relevant for various inflammatory and metabolic disorders.
Collapse
Affiliation(s)
- Wanling Huang
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510180, PR China
| | - Gaohong Fu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Yangeng Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Cheng Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Yilan Luo
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Qiaoqiao Yan
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
| | - Yang Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, PR China.
| | - Chengqiong Mao
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510180, PR China.
| |
Collapse
|