1
|
Shirley S, Ichise H, Di Natale V, Jin J, Wu C, Zou R, Zhang W, Fang Y, Zhang Y, Chen M, Peng S, Basu U, Que J, Huang Y. A vasculature-resident innate lymphoid cell population in mouse lungs. Nat Commun 2025; 16:3718. [PMID: 40253407 PMCID: PMC12009297 DOI: 10.1038/s41467-025-58982-1] [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: 09/29/2024] [Accepted: 04/08/2025] [Indexed: 04/21/2025] Open
Abstract
Tissue-resident immune cells such as innate lymphoid cells (ILC) are known to reside in the parenchymal compartments of tissues and modulate local immune protection. Here we use intravascular cell labeling, parabiosis and multiplex 3D imaging to identify a population of group 3 ILCs in mice that are present within the intravascular space of lung blood vessels (vILC3). vILC3s are distributed broadly in alveolar capillary beds from which inhaled pathogens enter the lung parenchyma. By contrast, conventional ILC3s in tissue parenchyma are enriched in lymphoid clusters in proximity to large veins. In a mouse model of pneumonia, Pseudomonas aeruginosa infection results in rapid vILC3 expansion and production of chemokines including CCL4. Blocking CCL4 in vivo attenuates neutrophil recruitment to the lung at the early stage of infection, resulting in prolonged inflammation and delayed bacterial clearance. Our findings thus define the intravascular space as a site of ILC residence in mice, and reveal a unique immune cell population that interfaces with tissue alarmins and the circulating immune system for timely host defense.
Collapse
Affiliation(s)
- Simon Shirley
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Hiroshi Ichise
- Lymphocyte Biology Section, Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Vincenzo Di Natale
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Jiacheng Jin
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Christine Wu
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Raymond Zou
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Wanwei Zhang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Yinshan Fang
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, USA
| | - Yingyu Zhang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Miao Chen
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Sophia Peng
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Uttiya Basu
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Jianwen Que
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, USA.
| | - Yuefeng Huang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
2
|
Ngambenjawong C, Ko H, Samad T, Pishesha N, Ploegh HL, Bhatia SN. Nanobody-Targeted Conditional Antimicrobial Therapeutics. ACS NANO 2025; 19:9958-9970. [PMID: 40044143 PMCID: PMC11924319 DOI: 10.1021/acsnano.4c16007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 03/19/2025]
Abstract
Conditional therapeutics that rely on disease microenvironment-specific triggers for activation are a promising strategy to improve therapeutic cargos. Among the investigated triggers, protease activity is used most often because of its dysregulation in several diseases. How to optimally fine-tune protease activation for different therapeutic cargos remains a challenge. Here, we designed nanobody-targeted conditional antimicrobial therapeutics to deliver a model therapeutic peptide and protein to the site of bacterial infection. We explored several parameters that influence proteolytic activation. We report the use of targeting nanobodies to enhance the activation of therapeutics that are otherwise activated inefficiently despite extensive optimization of the cleavable linker. Specifically, the pairing of Ly6G/C or ADAM10-targeting nanobodies with ADAM10-cleavable linkers improved activation via proximity-enabled reactivity. This study demonstrates a distinct role of active targeting in conditional therapeutic activation. More broadly, this optimization framework provides a guideline for the development of conditional therapeutics to treat various diseases in which protease activity is dysregulated.
Collapse
Affiliation(s)
- Chayanon Ngambenjawong
- Koch Institute
for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute
for Medical Engineering and Science, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- School of
Biomolecular Science and Engineering, Vidyasirimedhi
Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Henry Ko
- Koch Institute
for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute
for Medical Engineering and Science, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Tahoura Samad
- Koch Institute
for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute
for Medical Engineering and Science, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Novalia Pishesha
- Koch Institute
for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Division
of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Hidde L. Ploegh
- Program in
Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sangeeta N. Bhatia
- Koch Institute
for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute
for Medical Engineering and Science, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Howard
Hughes
Medical Institute, Cambridge, Massachusetts 02139, United States
- Department
of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Medicine, Brigham and Women’s
Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
- Broad
Institute
of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
3
|
Su Y, Feng C, Ye W, Xiao J, Meng Q, Yang X, Wang Y, Huang T, Lan L, Chen S, Ding Z, Su S, Wei S, Shan Q. Exploring the dynamic responses of group 3 innate lymphoid cells at different times in response to LPS challenge. Int Immunopharmacol 2025; 148:114162. [PMID: 39889415 DOI: 10.1016/j.intimp.2025.114162] [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: 11/24/2024] [Revised: 01/12/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Group 3 innate lymphoid cells (ILC3s) have clear roles in regulating mucosal immunity and tissue homeostasis in the intestine, though the immunological functions in lungs remain unclear. This study aimed to demonstrate the dynamic responses of ILC3s to acute inflammation upon LPS challenge. Microarray data and single-cell RNA sequencing (scRNA-seq) data obtained from the GEO database were combined to analyze the function of ILC3 subset, confirmed by flow cytometry assay and qRT-PCR. The gene enrichment analysis of intersected genes identified between microarray data in bacterial pneumonia and single-cell RNA sequencing of intestinal ILC3s were closely related to TNF-alpha effects on cytokine activity, cell motility and apoptosis pathway, indicating the possibility of intestinal ILC3s migration to the lung. Furthermore, the cellular landscapes of ILC3s in lung and intestine at different times after pulmonary infection exhibited varied ILC3 statuses. ILC3s in lung expanded a lot at 48 h while intestinal ILC3s decreased at 72 h response to LPS challenge, with higher expression of marked genes related to TNF-alpha effects on cytokine activity, cell motility and apoptosis pathway. The main findings in our study may serve as valuable resources for understanding the roles that ILC3s play upon LPS challenge, which may offer opportunities for translating ILC3s as therapeutic targets to regulate LPS-induced pulmonary inflammation.
Collapse
Affiliation(s)
- Ying Su
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Caixia Feng
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Wenyu Ye
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Juan Xiao
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Qi Meng
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Xia Yang
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Yongcai Wang
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Ting Huang
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture Guangxi Academy of Fishery Sciences Nanning China
| | - Liancheng Lan
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Sixing Chen
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Ziting Ding
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Shiqi Su
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Sumei Wei
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China
| | - Qingwen Shan
- Department of Pediatrics The First Affiliated Hospital of Guangxi Medical University/Difficult and Critical Illness Center Pediatric Clinical Medical Research Center of Guangxi Nanning China.
| |
Collapse
|
4
|
Xie Y, Shi Y, Wang L, Li C, Wu M, Xu J. Outer membrane vesicle contributes to the Pseudomonas aeruginosa resistance to antimicrobial peptides in the acidic airway of bronchiectasis patients. MedComm (Beijing) 2025; 6:e70084. [PMID: 39896756 PMCID: PMC11782972 DOI: 10.1002/mco2.70084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 09/29/2024] [Accepted: 11/23/2024] [Indexed: 02/04/2025] Open
Abstract
Pseudomonas aeruginosa is the predominant pathogen causing chronic infection in the airway of patients with bronchiectasis (BE), a chronic respiratory disease with high prevalence worldwide. Environmental factors are vital for bacterial successful colonization. Here, with sputa and bronchoalveolar lavage fluids, we determined that the concentration of airway antimicrobial peptide LL-37 and lactate was elevated in BE patients, especially in those infected with P. aeruginosa. The in vitro antibacterial assay revealed the bactericidal activity of LL-37 against the clinical P. aeruginosa isolates, which were dampened in the acidic condition. P. aeruginosa production of outer membrane vesicles (OMVs) enhanced in the lactate-adjusted acidic condition. Transcriptomic analysis suggested that OMVs induce the hyperproduction of the chemical compound 2-heptyl-4-quinolone (HHQ) in the bacterial population, which was verified by high-performance liquid chromatography. The positively charged HHQ interfered with the binding of LL-37 to bacterial cell membrane, potentiating the P. aeruginosa resistance to LL-37. To our knowledge, this is a new resistance mechanism of P. aeruginosa against antimicrobial peptides and may provide theoretical support for the development of new antibacterial therapies.
Collapse
Affiliation(s)
- Yingzhou Xie
- Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of MedicineTongji UniversityShanghaiChina
| | - Yi‐Han Shi
- Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of MedicineTongji UniversityShanghaiChina
| | - Le‐Le Wang
- Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of MedicineTongji UniversityShanghaiChina
| | - Cheng‐Wei Li
- Department of Pulmonary and Critical Care MedicineHuashan Hospital, Fudan UniversityShanghaiChina
| | - Min Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, and Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouChina
| | - Jin‐Fu Xu
- Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of MedicineTongji UniversityShanghaiChina
- Department of Respiratory and Critical Care MedicineHuadong Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
5
|
Shen X, Jin Z, Chen X, Wang Z, Yi L, Ou Y, Gong L, Zhu C, Xu G, Wang Y. Single-cell transcriptome atlas revealed bronchoalveolar immune features related to disease severity in pediatric Mycoplasma pneumoniae pneumonia. MedComm (Beijing) 2024; 5:e748. [PMID: 39399649 PMCID: PMC11471001 DOI: 10.1002/mco2.748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 10/15/2024] Open
Abstract
The mechanisms underlying protective immunity in mild Mycoplasma pneumoniae pneumonia (MPP) and the pathogenesis of severe MPP, characterized by dysregulated immune responses, remain unclear. Here, we performed single-cell RNA sequencing (scRNA-seq) to profile bronchoalveolar lavage fluid (BALF) samples from 13 healthy donors and 24 hospitalized pediatric patients with MPP, covering both mild and severe cases. Severe MPP patients exhibited high levels of exhausted T cells and M1-like macrophages, with the exhaustion of T cells attributed to persistent type I interferon signaling and inadequate assistance from CD4+ T cells. Significant cell-cell interactions between exhausted T cells and programmed death-ligand 1+ (PD-L1+) macrophages were detected in severe patients, potentially mediated through inhibitor molecules (e.g., PD1) and their receptors (e.g., PD-L1), as well as human leukocyte antigen class I molecules and their receptors (e.g., KLRC1/D2), resulting in the dysfunction of anti-MP immune responses. Mild MPP patients were featured by an increased abundance of neutrophils, coupled with enhanced activation, contributing to protective immunity. Together, our study provides a detailed characterization of the BALF immune landscape in MPP patients, revealing distinct immune characteristics between mild and severe cases, which offers a valuable resource for understanding MPP immunopathogenesis and formulating effective therapeutic strategies.
Collapse
Affiliation(s)
- Xiantao Shen
- State Key Laboratory of Environment Health (Incubation)Key Laboratory of Environment and HealthMinistry of EducationKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionSchool of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhengjiang Jin
- Department of Clinical LaboratoryMaternal and Child Health Hospital of Hubei ProvinceTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaomin Chen
- Department of Disinfection and Pest ControlWuhan Center for Disease Control & PreventionWuhanChina
| | - Zhenhui Wang
- Department of Clinical LaboratoryMaternal and Child Health Hospital of Hubei ProvinceTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lu Yi
- Department of Clinical LaboratoryMaternal and Child Health Hospital of Hubei ProvinceTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yangwei Ou
- Department of RadiologyMaternal and Child Health Hospital of Hubei ProvinceTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lin Gong
- State Key Laboratory of Environment Health (Incubation)Key Laboratory of Environment and HealthMinistry of EducationKey Laboratory of Environment and Health (Wuhan)Ministry of Environmental ProtectionSchool of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Disinfection and Pest ControlWuhan Center for Disease Control & PreventionWuhanChina
| | - Chengliang Zhu
- Department of Clinical LaboratoryInstitute of Translational MedicineRenmin Hospital of Wuhan UniversityWuhanChina
| | - Guogang Xu
- Health Management InstituteThe Second Medical Center & National Clinical Research Center for Geriatric DiseasesChinese PLA General HospitalBeijingChina
| | - Yi Wang
- Experimental Research CenterCapital Institute of PediatricsBeijingChina
| |
Collapse
|
6
|
Xu W, Ye J, Cao Z, Zhao Y, Zhu Y, Li L. Glucocorticoids in lung cancer: Navigating the balance between immunosuppression and therapeutic efficacy. Heliyon 2024; 10:e32357. [PMID: 39022002 PMCID: PMC11252876 DOI: 10.1016/j.heliyon.2024.e32357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
Glucocorticoids (GCs), a class of hormones secreted by the adrenal glands, are released into the bloodstream to maintain homeostasis and modulate responses to various stressors. These hormones function by binding to the widely expressed GC receptor (GR), thereby regulating a wide range of pathophysiological processes, especially in metabolism and immunity. The role of GCs in the tumor immune microenvironment (TIME) of lung cancer (LC) has been a focal point of research. As immunosuppressive agents, GCs exert a crucial impact on the occurrence, progression, and treatment of LC. In the TIME of LC, GCs act as a constantly swinging pendulum, simultaneously offering tumor-suppressive properties while diminishing the efficacy of immune-based therapies. The present study reviews the role and mechanisms of GCs in the TIME of LC.
Collapse
Affiliation(s)
| | | | - Zhendong Cao
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| | - Yupei Zhao
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| | - Yimin Zhu
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| | - Lei Li
- Department of Respiration, The Second Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (Jiangsu Second Hospital of Traditional Chinese Medicine), Nanjing, Jiangsu, 210017, China
| |
Collapse
|
7
|
Li R, Li J, Zhou X. Lung microbiome: new insights into the pathogenesis of respiratory diseases. Signal Transduct Target Ther 2024; 9:19. [PMID: 38228603 PMCID: PMC10791971 DOI: 10.1038/s41392-023-01722-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/25/2023] [Accepted: 11/22/2023] [Indexed: 01/18/2024] Open
Abstract
The lungs were long thought to be sterile until technical advances uncovered the presence of the lung microbial community. The microbiome of healthy lungs is mainly derived from the upper respiratory tract (URT) microbiome but also has its own characteristic flora. The selection mechanisms in the lung, including clearance by coughing, pulmonary macrophages, the oscillation of respiratory cilia, and bacterial inhibition by alveolar surfactant, keep the microbiome transient and mobile, which is different from the microbiome in other organs. The pulmonary bacteriome has been intensively studied recently, but relatively little research has focused on the mycobiome and virome. This up-to-date review retrospectively summarizes the lung microbiome's history, composition, and function. We focus on the interaction of the lung microbiome with the oropharynx and gut microbiome and emphasize the role it plays in the innate and adaptive immune responses. More importantly, we focus on multiple respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), fibrosis, bronchiectasis, and pneumonia. The impact of the lung microbiome on coronavirus disease 2019 (COVID-19) and lung cancer has also been comprehensively studied. Furthermore, by summarizing the therapeutic potential of the lung microbiome in lung diseases and examining the shortcomings of the field, we propose an outlook of the direction of lung microbiome research.
Collapse
Affiliation(s)
- Ruomeng Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Xikun Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
8
|
Wang X, Liu M, Yu C, Li J, Zhou X. Biofilm formation: mechanistic insights and therapeutic targets. MOLECULAR BIOMEDICINE 2023; 4:49. [PMID: 38097907 PMCID: PMC10721784 DOI: 10.1186/s43556-023-00164-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
Biofilms are complex multicellular communities formed by bacteria, and their extracellular polymeric substances are observed as surface-attached or non-surface-attached aggregates. Many types of bacterial species found in living hosts or environments can form biofilms. These include pathogenic bacteria such as Pseudomonas, which can act as persistent infectious hosts and are responsible for a wide range of chronic diseases as well as the emergence of antibiotic resistance, thereby making them difficult to eliminate. Pseudomonas aeruginosa has emerged as a model organism for studying biofilm formation. In addition, other Pseudomonas utilize biofilm formation in plant colonization and environmental persistence. Biofilms are effective in aiding bacterial colonization, enhancing bacterial resistance to antimicrobial substances and host immune responses, and facilitating cell‒cell signalling exchanges between community bacteria. The lack of antibiotics targeting biofilms in the drug discovery process indicates the need to design new biofilm inhibitors as antimicrobial drugs using various strategies and targeting different stages of biofilm formation. Growing strategies that have been developed to combat biofilm formation include targeting bacterial enzymes, as well as those involved in the quorum sensing and adhesion pathways. In this review, with Pseudomonas as the primary subject of study, we review and discuss the mechanisms of bacterial biofilm formation and current therapeutic approaches, emphasizing the clinical issues associated with biofilm infections and focusing on current and emerging antibiotic biofilm strategies.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ming Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chuanjiang Yu
- Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Xikun Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|