Research paper
Specific discrimination and efficient elimination of gram-positive bacteria by an aggregation-induced emission-active ruthenium (II) photosensitizer

https://doi.org/10.1016/j.ejmech.2023.115249Get rights and content

Highlights

  • A multifunctional ruthenium (II) polypyridine complex with aggregation-induced emission (AIE) characteristic was synthesized.

  • The selective discrimination and imaging of gram-positive bacteria was with Ru2.

  • Ru2 under light irradiation had the best antibacterial activity in vitro and in vivo.

  • Ru2 altered Gram-positive bacterial gene expression and successfully applied to S. aureus-infected wound treatment.

Abstract

The infections caused by Gram-positive bacteria (G+) have seriously endangered public heath due to their high morbidity and mortality. Therefore, it is urgent to develop a multifunctional system for selective recognition, imaging and efficient eradication of G+. Aggregation-induced emission materials have shown great promise for microbial detection and antimicrobial therapy. In this paper, a multifunctional ruthenium (II) polypyridine complex Ru2 with aggregation-induced emission (AIE) characteristic, was developed and used for selective discrimination and efficient extermination of G+ from other bacteria with unique selectivity. The selective G+ recognition benefited from the interaction between lipoteichoic acids (LTA) and Ru2. Accumulation of Ru2 on the G+ membrane turned on its AIE luminescence and allowed specific G+ staining. Meanwhile, Ru2 under light irradiation also possessed robust antibacterial activity for G+ in vitro and in vivo antibacterial experiments. To the best of our knowledge, Ru2 is the first Ru-based AIEgen photosensitizer for simultaneous dual applications of G+ detection and treatment, and inspires the development of promising antibacterial agents in the future.

Introduction

Bacterial infections caused by Gram-positive bacteria (G+) remain a leading cause of morbidity and mortality in humans [1,2]. G+ causes a variety of infections that include skin and soft tissue infections [3], surgical site infections [4], bloodstream infections [5] and pneumonia infections [6], which are closely related to endocarditis, pneumonia, arthritis, osteomyelitis, and sepsis [7]. Notably, some G+ existing in tumors may inhibit the curative efficacy and promote chemoresistance of chemotherapy drugs [8,9]. Therefore, it is of great significance to develop new antibacterial agents, especially those that can accurately identify and kill G+ [10,11].

The main methods for G+ identification include the Gram staining [12], polymerase chain reaction (PCR) [13], nucleic acid-related methods [14], immunological techniques [15], and surface enhanced Raman spectroscopy (SERS) [16], which have generally long running time, low accuracy, complicated operation and instrumental dependence [10]. Compared to these aforementioned methods, fluorescent detection techniques are an alternative approach due to their high sensitivity, fast response, non-invasiveness, and direct observation, and have been rapidly developed and applied for bacteria detection and identification [17]. Additionally, the effective killing of G+ based on accurate identification was indeed the ultimate goal to combat G+ for humans. Antibiotics are the most widely accepted bactericide at present. However, the occurrence of multidrug-resistant strains associated with the abuse of antibiotics narrows the clinical option of traditional antibiotics [18,19]. Antimicrobial photodynamic therapy (APDT) has been considered one of the most promising methods to overcome bacterial resistance [[20], [21], [22]]. Through irradiation, the activated photosensitizers (PSs) can transform oxygen molecules to reactive oxygen species (ROS), mainly singlet oxygen (1O2), which can disrupt bacterial cell membranes and irreversibly damage biomolecules, such as nucleic acids, lipids, and proteins, eventually leading to bacterial cell death [23]. Besides, PSs have been utilized in fluorescence image-guided antibacterial studies [22]. However, most of the PSs are hydrophobic and tend to aggregate when interacting with bacteria, leading to fluorescence quenching and low efficiency of ROS generation, which attenuated the imaging performance and antibacterial activity [24,25]. Aggregation-induced emission luminogens (AIEgens) possessed enhanced fluorescence intensity and high 1O2 generation efficiency in the aggregated state through the intramolecular rotation/motion restriction, which could be a potential solution against bacteria [26]. Up to now, some elegant AIEgens have been developed for bacterial detection or ablation [[27], [28], [29]]. Among the multifarious AIEgens, heavy metal-based AIEgens gained wide interest due to their rich luminescent properties, and straightforward synthesis routes, high photo-induced ROS yields, which made them promising alternatives to organic AIEgens for the bacteria detection, imaging and therapy [29]. In this field, a few rare examples of AIE-derived transition metal complexes were those of Ir(III) [30], Pt(II) [31], Zn(II) [32] and Au(I) [33]. Such complexes have been used as probes for the detection and eradication of bacteria. However, the metal ions employed in these complexes were not abundant and were often absent from the selective discrimination ability of bacteria. It was, therefore, essential to develop other transition metal complexes that can discriminate and have pharmacologic activity for bacteria.

The cell wall of G+ contains multiple thick peptidoglycans and teichoic acid layers (LTA), while the cell wall of G is composed of 2–3 layers of peptidoglycan surrounded by a second outer membrane containing lipopolysaccharide (LPS) and lipoprotein. As a result, the specific outer wall structures of bacteria play a crucial role in determining the different binding interactions of metal-based AIEgens with bacteria and enable us to discriminate them by AIEgens [29].

Herein, a series of multifunctional ruthenium-based AIE probes Ru1-3 were designed and synthesized; their ability of imaging and efficient eradication of G+ were also investigated (Fig. 1). Ru2 could rapidly distinguish G+ through specific binding to the LTA in the G+ cell wall without an extra targeting group. More importantly, Ru2 could selectively discriminate G+ even in the presence of G. Besides, the effective growth inhibition of G+ upon Ru2 treatment under light irradiation was validated in vitro and in vivo. To the best of our knowledge, this was the first Ru(II) complex for specific discrimination, imaging, and sterilization of G+. This study offered useful guidance for designing the next-generation antibacterial agents.

Section snippets

Ruthenium(II)-based AIEgens synthesis

A series of cationic ruthenium(II) polypyridine complexes (Ru1−3) were designed and synthesized to serve as antibacterial agents (Fig. 1). Detailed synthesis and characterizations of Ru1−3 were shown in the Supporting Information (Fig. S1−S11, S12A−C). The basic photophysical properties of Ru1−3 (10 μM in PBS, containing 1% DMSO) were characterized and presented in Fig. S12D−E and Table S1. Ru1 showed relatively intense photoluminescence with quantum yields (ФPL) of 0.09 compared to Ru(bpy)32+

Conclusions

In summary, we prepared a multifunctional probe Ru2 with AIE characteristics for selective imaging and killing G+ including MRSA strains. Due to its AIE property and strong binding ability with G+ cell wall, the cationic Ru2 could selectively stain G+. Moreover, Ru2 could selectively discriminate G+ in the presence of G by a washing-free staining procedure. Both in vitro and in vivo antibacterial experiments manifested that Ru2 upon light irradiation could effectively suppress G+ growth, and

Credit author statement

Mengling Liu and Wenzhu Song performed the experiments and wrote the manuscript. Peipei Deng analyzed the results and drawn the figures. Yue Yu drawn the scheme. Shuli Nong and Xianpeng Zhang assisted the in vivo therapeutic evaluation. Li Xu and Guanying Li critically discussed the results and reviewed the manuscript. Li Xu supervised the project and interpreted the data. The paper was discussed and reviewed by all authors.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

This work was supported by the Science and Technology Planning Project of Guangzhou (No. 202002030089), the Key Projects of Social Welfare and Basic Research of Zhongshan City (2021B2007), the National Science Foundation of China (No. 31971314, 22107087), special funds of key disciplines construction from Guangdong and Zhongshan cooperating, partially supported by the “Yong Talent Support Plan” of Xi'an Jiaotong University.

References (49)

  • F.A. Manian et al.

    Surgical site infections associated with methicillin-resistant staphylococcus aureus: do postoperative factors play a role?

    Clin. Infect. Dis.

    (2003)
  • S.K. Choi et al.

    Dendrimer-based multivalent vancomycin nanoplatform for targeting the drug-resistant bacterial surface

    ACS Nano

    (2013)
  • T.C. Yu et al.

    Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy

    Cell

    (2017)
  • M. Kang et al.

    Evaluation of structure–function relationships of aggregation-induced emission luminogens for simultaneous dual applications of specific discrimination and efficient photodynamic killing of Gram-positive bacteria

    J. Am. Chem. Soc.

    (2019)
  • T. Dai et al.

    An AIEgen as an intrinsic antibacterial agent for light-up detection and inactivation of intracellular Gram-positive bacteria

    Adv. Healthcare Mater.

    (2021)
  • D. Claus

    A standardized Gram staining procedure

    World J. Microbiol. Biotechnol.

    (1992)
  • H.Y. Kwon et al.

    Development of a universal fluorescent probe for Gram-positive bacteria

    Angew. Chem., Int. Ed.

    (2019)
  • R.T. Ranasinghe et al.

    Detecting RNA base methylations in single cells by in situ hybridization

    Nat. Commun.

    (2018)
  • C.R. Bertozzi et al.

    Antibody targeting to bacterial cells using receptor-specific ligands

    J. Am. Chem. Soc.

    (1992)
  • A.K. Boardman et al.

    Rapid detection of bacteria from blood with surface-enhanced Raman spectroscopy

    Anal. Chem.

    (2016)
  • X. Feng et al.

    Recent progress of aggregation-induced emission luminogens (AIEgens) for bacterial detection and theranostics

    Mater. Chem. Front.

    (2021)
  • Y. Qiao et al.

    Treatment of MRSA-infected osteomyelitis using bacterial capturing, magnetically targeted composites with microwave-assisted bacterial killing

    Nat. Commun.

    (2020)
  • C.A. Arias et al.

    Antibiotic-resistant bugs in the 21st century-a clinical super-challenge

    N. Engl. J. Med.

    (2009)
  • T. Zhou et al.

    An AIE-active conjugated polymer with high ROS-generation ability and biocompatibility for efficient photodynamic therapy of bacterial infections

    Angew. Chem., Int. Ed.

    (2020)
  • Cited by (6)

    1

    These authors contributed equally to this work.

    View full text