1
|
Luo L, Li Q, Xing C, Li C, Pan Y, Sun H, Yu X, Wen K, Shen J, Wang Z. Antibody-based therapy: An alternative for antimicrobial treatment in the post-antibiotic era. Microbiol Res 2025; 290:127974. [PMID: 39577369 DOI: 10.1016/j.micres.2024.127974] [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: 10/01/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
The consecutive growth of antimicrobial resistance and the spread of resistance genes worldwide, especially the emergence of superbugs, have made traditional antibiotic-based treatments inadequate to fight bacterial infections. Therefore, new therapeutic modalities for bacterial infections are urgently needed. Antibodies are considered to be an effective alternative to antibiotics. The emergence and advancement of technologies such as hybridoma, antibody purification, transgenic mice, phage display, and protein engineering have enabled the production of large quantities of humanized antibodies with high purity and affinity. Antibodies has achieved remarkable achievements in the field of medicine in the past decades. Antibody-based therapy is expected to be an effective way to treat drug-resistant bacterial infections in the post-antibiotic era due to its merits of high specificity, which leads to no selective pressure on non-target bacteria and could cooperate with antibiotics to enhance the antimicrobial effect. This review first introduces the mechanism of action of antibodies against bacterial infections, then summarizes the reported antimicrobial antibodies according to different targets, discusses the advantages and limitations of the antibody-based therapy for antimicrobial treatment, and finally, the perspectives of antimicrobial antibodies developing have been prospected, providing a reference for the development of new antimicrobial antibodies.
Collapse
Affiliation(s)
- Liang Luo
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China; Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, PR China
| | - Qing Li
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Chen Xing
- Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, PR China
| | - Chenglong Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, PR China
| | - Yantong Pan
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China; Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, PR China
| | - He Sun
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Xuezhi Yu
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Kai Wen
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China
| | - Zhanhui Wang
- National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, PR China; Technology Innovation Center for Food Safety Surveillance and Detection (Hainan), Sanya Institute of China Agricultural University, Sanya 572025, PR China.
| |
Collapse
|
2
|
Li M, Abouelfetouh MM, Salah E, Kiani FA, Nan S, Ding M, Ding Y. Chicory supplementation improves growth performance in juvenile ostriches potentially by attenuating enteritis. Front Vet Sci 2024; 11:1432269. [PMID: 39376909 PMCID: PMC11457291 DOI: 10.3389/fvets.2024.1432269] [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: 05/16/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
Introduction Enteritis and dysbiosis are the major causes of high morbidity and mortality of juvenile ostriches. Chicory (CC) has been proven to have excellent antioxidant, anti-inflammatory, and antibacterial activities. However, it's unclear whether CC could improve the survival rate of juvenile ostriches by relieving enteritis and correcting dysbiosis. Materials and methods South African ostrich hatchlings (Struthio camelus domesticus) were fed with and without a CC-supplemented diet, and the body weight gain and mortality were compared over 4 months of age. Fresh fecal samples of clinically healthy ostriches were collected, and 16S DNAs were analyzed. Moreover, ostrich chicks with LPS-induced enteritis were fed with different dosages (0, 20, 40, and 80 mg/kg) of chicoric acid (CA), a major bioactive component of CC, for five consecutive days. The expression levels of tight junction (TJ)-related proteins and inflammatory mediators in the ilea were detected with western blot and immunofluorescence. Results The ostrich chicks fed on the CC-supplemented diet began to increase in weight at the 1st month of age and became remarkably heavier at the fourth month (p < 0.01) compared with those fed on the non-CC-supplemented diet. Additionally, the mortality percentage was lower in the chicks fed on the CC-supplemented diet than those fed on the non-CC-supplemented diet (19% vs. 36%, respectively). The diet with the CC supplementation significantly increased the abundance of Phascolactobacteria (linear discriminant analysis; LDA >4) and Bacteroidota (26.7% vs. 17.7%, respectively) as well as decreased the enrichment of Clostridium (5.0% vs. 9.1%, respectively) in the ostrich ilea compared to the diet without CC. The supplementation of CA at a dose of 80 mg/kg significantly increased the expression level of ZO-1 and claudin-3 (p < 0.0001) and suppressed the levels of IL-1β, IL-6, and TNF-α (p < 0.0001) in ostriches with LPS-induced ileitis. Conclusion Our results substantiate that CC or CA supplementation in a diet could effectively improve growth performance and reduce mortality in juvenile ostriches via modulating the gut microbiota and attenuating enteritis.
Collapse
Affiliation(s)
- Meng Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Henan Jinlu Special Breeding Farm, Zhengzhou, China
| | - Mahmoud M. Abouelfetouh
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Egypt
| | - Eman Salah
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Egypt
| | - Faisal Ayub Kiani
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Bahauddin Zakariyah University, Multan, Pakistan
| | - Sha Nan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Mingxing Ding
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yi Ding
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
3
|
Liu B, Cao J, Hong B, You H, Li T, Yu Z, Li D, Liang B, Gan N. A microfluidic chip platform based on Pt nanozyme and magnetized phage composite probes for dual-mode detecting and imaging pathogenic bacteria viability. Talanta 2024; 275:126067. [PMID: 38640522 DOI: 10.1016/j.talanta.2024.126067] [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: 02/01/2024] [Revised: 03/30/2024] [Accepted: 04/05/2024] [Indexed: 04/21/2024]
Abstract
The detection of pathogen viability is critically important to evaluate its infectivity. In the study, an integrated microfluidic chip based on dual-mode analytical strategy was developed to rapidly realize detection of bacteria activity (with Salmonella typhimurium, S.T, as a model analyte). Firstly, the composite probes, including deactivated phage modified magnetic beads and nano Pt-antimicrobial peptide (AMP) which can specifically recognize Gram-negative bacteria as nanozyme were prepared. When the composite probes are introduced into the chip together with target bacteria, after enrichment, oscillating and magnetic separation, they will conjugate with S.T and produce a magnetic sandwich complex. The complex can catalyze tetramethylbenzidine (TMB)-H2O2 to produce visible colorimetric signals which is correspondent to the total S.T content. Simultaneously, PtNPs in the complex can produce hydroxyl radical oxidation (∙OH) by decomposing H2O2. Under the synergistic action of ∙OH and AMP, the captured live S.T can be lysed to release ATP and emit bioluminescence signals which corresponds to the live S.T concentration. Therefore, the chip can simultaneously detect and image S.T at different viability in one test. The dual-mode assay demonstrated high sensitivity (≤33 CFU/mL), high specificity (identifying strain), signal amplification (5 folds) and short time (≤40min). The chip array can detect four samples in one test and exhibited advantages of high-integration, -sensitivity, -specificity and miniaturization, which are suitable to rapidly detect and image pathogen's viability in trace level. The replacement of phage probes can detect other bacteria. It has a wide prospect in pathogens screening.
Collapse
Affiliation(s)
- Bailu Liu
- School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang province, 315211, China
| | - Jingya Cao
- School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang province, 315211, China
| | - Binxin Hong
- School of Marine Science, Ningbo University, Ningbo, 315211, China
| | - Hang You
- School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang province, 315211, China
| | - Tianhua Li
- School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang province, 315211, China
| | - Zhenzhong Yu
- School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang province, 315211, China
| | - Dengfeng Li
- School of Marine Science, Ningbo University, Ningbo, 315211, China
| | - Baihui Liang
- Healthy & Intelligent Kitchen Engineering Research Center of Zhejiang Province, Ningbo, 315336, China; Ningbo Fotile Kitchenware Co., Ltd., Ningbo, Zhejiang 315336, China.
| | - Ning Gan
- School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang province, 315211, China.
| |
Collapse
|
4
|
Chen H, Yang T, Xu Y, Liang B, Liu X, Cai Y. Anti-inflammatory and immunoregulatory effects of colistin sulphate on human PBMCs. J Cell Mol Med 2024; 28:e18322. [PMID: 38661452 PMCID: PMC11044820 DOI: 10.1111/jcmm.18322] [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: 11/19/2023] [Revised: 03/06/2024] [Accepted: 04/03/2024] [Indexed: 04/26/2024] Open
Abstract
In previous studies, CST has been identified as having an immunostimulatory effect on Caenorhabditis elegans and macrophage of rats. Here, we further investigated its immunomodulatory effects on human peripheral blood mononuclear cells (PBMCs). LPS-stimulated PBMCs inflammatory model was established. Flow cytometry was applied to measure phagocytosis of PBMCs. Cytokine mRNA and protein expression levels of LPS-stimulated PBMCs with or without CST were measured by qRT-PCR and ELISA. The transcriptomic profile of CST-treated PBMCs was investigated by RNA-sequencing. Gene Ontology (GO) and Kyoto Encylopedia of Genes and Genomes (KEGG) were applied to find potential signalling pathways. PBMCs showed a significant increase in phagocytic activity at 6 h after being incubated with CST at the concentration of 10 μg/mL. In the presence of LPS, CST maintained and promoted the expression of TNF-α and chemokine CCL24. The content of pro-inflammatory cytokines, such as IL-1β, IL-6 and IFN-γ, which were released from LPS-stimulated PBMCs, was reduced by CST at 6 h. Anti-inflammatory cytokines, such as IL-4, IL-13 and TGF-β1, were significantly increased by CST at 24 h. A total of 277 differentially expressed immune-related genes (DEIRGs) were detected and cytokine-cytokine receptor interaction was highly enriched. CST presented obvious anti-inflammatory and immunoregulatory effects in LPS-induced PBMCs inflammatory model not only by improving the ability of PBMCs to clear pathogens but also by decreasing pro-inflammatory cytokines and increasing anti-inflammatory cytokines. And the mechanism may be related to cytokine-cytokine receptor interaction.
Collapse
Affiliation(s)
- Huiling Chen
- Department of PharmacyCenter of Medicine Clinical Research, Medical Supplies Center, PLA General HospitalBeijingChina
- Department of PharmacyZigong Fourth People's HospitalZigongChina
| | - Tianli Yang
- Department of PharmacyCenter of Medicine Clinical Research, Medical Supplies Center, PLA General HospitalBeijingChina
- Medical School of Chinese PLAGraduate School of Chinese PLA General HospitalBeijingChina
| | - Yiran Xu
- Department of PharmacyCenter of Medicine Clinical Research, Medical Supplies Center, PLA General HospitalBeijingChina
- The Second Naval Hospital of Southern Theater Command of PLASanyaChina
| | - Beibei Liang
- Department of PharmacyCenter of Medicine Clinical Research, Medical Supplies Center, PLA General HospitalBeijingChina
| | - Xianyong Liu
- Medical School of Chinese PLAGraduate School of Chinese PLA General HospitalBeijingChina
- Department of Thoracic SurgeryThe First Medical Center, PLA General HospitalBeijingChina
| | - Yun Cai
- Department of PharmacyCenter of Medicine Clinical Research, Medical Supplies Center, PLA General HospitalBeijingChina
| |
Collapse
|
5
|
Brandenburg K, Ferrer-Espada R, Martinez-de-Tejada G, Nehls C, Fukuoka S, Mauss K, Weindl G, Garidel P. A Comparison between SARS-CoV-2 and Gram-Negative Bacteria-Induced Hyperinflammation and Sepsis. Int J Mol Sci 2023; 24:15169. [PMID: 37894850 PMCID: PMC10607443 DOI: 10.3390/ijms242015169] [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: 08/28/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Sepsis is a life-threatening condition caused by the body's overwhelming response to an infection, such as pneumonia or urinary tract infection. It occurs when the immune system releases cytokines into the bloodstream, triggering widespread inflammation. If not treated, it can lead to organ failure and death. Unfortunately, sepsis has a high mortality rate, with studies reporting rates ranging from 20% to over 50%, depending on the severity and promptness of treatment. According to the World Health Organization (WHO), the annual death toll in the world is about 11 million. One of the main toxins responsible for inflammation induction are lipopolysaccharides (LPS, endotoxin) from Gram-negative bacteria, which rank among the most potent immunostimulants found in nature. Antibiotics are consistently prescribed as a part of anti-sepsis-therapy. However, antibiotic therapy (i) is increasingly ineffective due to resistance development and (ii) most antibiotics are unable to bind and neutralize LPS, a prerequisite to inhibit the interaction of endotoxin with its cellular receptor complex, namely Toll-like receptor 4 (TLR4)/MD-2, responsible for the intracellular cascade leading to pro-inflammatory cytokine secretion. The pandemic virus SARS-CoV-2 has infected hundreds of millions of humans worldwide since its emergence in 2019. The COVID-19 (Coronavirus disease-19) caused by this virus is associated with high lethality, particularly for elderly and immunocompromised people. As of August 2023, nearly 7 million deaths were reported worldwide due to this disease. According to some reported studies, upregulation of TLR4 and the subsequent inflammatory signaling detected in COVID-19 patients "mimics bacterial sepsis". Furthermore, the immune response to SARS-CoV-2 was described by others as "mirror image of sepsis". Similarly, the cytokine profile in sera from severe COVID-19 patients was very similar to those suffering from the acute respiratory distress syndrome (ARDS) and sepsis. Finally, the severe COVID-19 infection is frequently accompanied by bacterial co-infections, as well as by the presence of significant LPS concentrations. In the present review, we will analyze similarities and differences between COVID-19 and sepsis at the pathophysiological, epidemiological, and molecular levels.
Collapse
Affiliation(s)
- Klaus Brandenburg
- Brandenburg Antiinfektiva, c/o Forschungszentrum Borstel, Leibniz-Lungenzentrum, Parkallee 10, 23845 Borstel, Germany; (K.B.); (K.M.)
| | - Raquel Ferrer-Espada
- Department of Microbiology, University of Navarra, IdiSNA (Navarra Institute for Health Research), Irunlarrea 1, E-31008 Pamplona, Spain;
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Guillermo Martinez-de-Tejada
- Department of Microbiology, University of Navarra, IdiSNA (Navarra Institute for Health Research), Irunlarrea 1, E-31008 Pamplona, Spain;
| | - Christian Nehls
- Forschungszentrum Borstel, FG Biophysik, Parkallee 10, 23845 Borstel, Germany;
| | - Satoshi Fukuoka
- National Institute of Advanced Industrial Science and Technology (AIST), Takamatsu 761-0395, Japan;
| | - Karl Mauss
- Brandenburg Antiinfektiva, c/o Forschungszentrum Borstel, Leibniz-Lungenzentrum, Parkallee 10, 23845 Borstel, Germany; (K.B.); (K.M.)
- Sylter Klinik Karl Mauss, Dr.-Nicolas-Strasse 3, 25980 Westerland (Sylt), Germany
| | - Günther Weindl
- Pharmazeutisches Institut, Abteilung Pharmakologie und Toxikologie, Universität Bonn, Gerhard-Domagk-Str. 3, 53121 Bonn, Germany;
| | - Patrick Garidel
- Physikalische Chemie, Martin-Luther-Universität Halle-Wittenberg, 06108 Halle (Saale), Germany
| |
Collapse
|
6
|
Xiao S, Cao C, Ming T, Cao Y, Yu Z, Gan N. Simultaneous and rapid screening of live and dead E. coli O157:H7 with three signal outputs: An all-in-one biosensor using phage-apoferritin@CuO 2 signal tags on MXenes-modified electrode platform. JOURNAL OF HAZARDOUS MATERIALS 2023; 458:131875. [PMID: 37343409 DOI: 10.1016/j.jhazmat.2023.131875] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023]
Abstract
Simultaneous detection of live and dead bacteria is a huge challenge for food safety. To solve this issue, an all-in-one biosensor for bacteria was developed using the phage-apoferritin@CuO2 (phage-Apo@CP) probe on an antimicrobial peptide (AMP)/MXenes-modified detection platform. With the specific recognition of AMP and phage-Apo@CP, the biosensor for the target Escherichia coli O157:H7 (E. coli O157:H7) presented multi-mode (bioluminescent, colorimetric, and electrochemical) signals to simultaneously measure live and dead bacteria. The bioluminescent signal caused by the adenosine triphosphate (ATP) from the bacteria was used to quantify live bacteria. The colorimetric and voltammetric signals triggered by ·OH and Cu2+ from the probe with the assistance of acid could rapidly screen and quantitative determination of total E. coli O157:H7 concentration. Thus, the dead one was obtained according to the total and live ones. All three signals could be mutually corrected to improve the accuracy. The biosensor was successfully used for on-site measurement of live and dead E. coli O157:H7 in food samples with the limit of detection of 30 CFU/mL for live ones and 6 CFU/mL for total bacteria within 50 min. This work presents a novel pathway for rapid and simultaneous quantification of both live and dead bacteria.
Collapse
Affiliation(s)
- Shu Xiao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China
| | - Cong Cao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China
| | - Tinghong Ming
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Yuting Cao
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China
| | - Zhenzhong Yu
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
| | - Ning Gan
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
7
|
Ji X, Yang X, Shi C, Guo D, Wang X, Messina JM, Meng Q, Urao N, Cooney R, Luo J. Functionalized core-shell nanogel scavenger for immune modulation therapy in sepsis. ADVANCED THERAPEUTICS 2022; 5:2200127. [PMID: 36590645 PMCID: PMC9797201 DOI: 10.1002/adtp.202200127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Indexed: 01/05/2023]
Abstract
Sepsis is a complex, life-threatening hyperinflammatory syndrome associated with organ failure and high mortality due to lack of effective treatment options. Here we report a core-shell hydrogel nanoparticle with the core functionalized with telodendrimer (TD) nanotrap (NT) to control hyperinflammation in sepsis. The combination of multi-valent charged and hydrophobic moieties in TD enables effective binding with biomolecules in NT. The higher crosslinking in the shell structure of nanogel excludes the abundant large serum proteins and allows for size-selectivity in scavenging the medium-sized septic molecules (10-30 kDa), e.g., lipopolysaccharides (LPS, a potent endotoxin in sepsis), thus reducing cytokine production. At the same time, the core-shell TD NT nanogel captures the over-flowing proinflammatory cytokines effectively both in vitro and in vivo from biological fluids to further control hyperinflammation. Intraperitoneal injection of core-shell TD NT nanogel effectively attenuates NF-κB activation and cytokine production in LPS-induced septic mouse models. These results indicate the potential applications of the injectable TD NT core-shell nanogel to attenuate local or systemic inflammation.
Collapse
Affiliation(s)
- Xiaotian Ji
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Xiguang Yang
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Changying Shi
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Dandan Guo
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Xiaojing Wang
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Jennifer M Messina
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Qinghe Meng
- Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Norifumi Urao
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
- Upstate Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Robert Cooney
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
- Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
- Upstate Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| | - Juntao Luo
- Department of Pharmacology, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
- Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
- Upstate Cancer Center, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
- Upstate Sepsis Interdisciplinary Research Center, State University of New York Upstate Medical University, Syracuse, NY 13210, United States
| |
Collapse
|
8
|
Baicalin Modulates Inflammatory Response of Macrophages Activated by LPS via Calcium-CHOP Pathway. Cells 2022; 11:cells11193076. [PMID: 36231038 PMCID: PMC9563021 DOI: 10.3390/cells11193076] [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: 08/15/2022] [Revised: 09/18/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Studies on natural products that can alleviate the inflammatory response of macrophages caused by endotoxin (lipopolysaccharide) continue. This study investigated the anti-inflammatory activity of baicalin related to macrophage activation caused by lipopolysaccharide (LPS). Baicalin is a flavone glycoside found in plants such as Scutellaria baicalensis and Scutellaria lateriflora belonging to the genus Scutellaria. The multiplex cytokine assay (MCA), Griess reagent assay, fluo-4 calcium assay, dihydrorhodamine 123 (DHR123) assay, quantitative RT-PCR, and flow cytometry were performed using RAW 264.7 mouse macrophages. The MCA revealed that baicalin significantly decreased the production of interleukin (IL)-6, granulocyte colony-stimulating factor (G-CSF), vascular endothelial growth factor (VEGF), macrophage inflammatory protein (MIP)-1α, MIP-1β, MIP-2, and RANTES in LPS-stimulated RAW 264.7 macrophages at concentrations of 10, 25, and 50 μM. The DHR123 assay showed that baicalin significantly inhibited reactive oxygen species generation in LPS-stimulated RAW 264.7 macrophages. Flow cytometry revealed that baicalin significantly reduced the levels of phosphorylated p38 MAPK and Fas in LPS-stimulated RAW 264.7 macrophages. Baicalin also inhibited the mRNA expression levels of inflammatory genes such as Chop, Fas, Nos2, Ptgs2, Stat1, c-Jun, c-Fos, and At1a. The IC50 values of baicalin for IL-6, TNF-α, G-CSF, VEGF, interferon gamma-induced protein 10 (IP-10), leukemia inhibitory factor (LIF), lipopolysaccharide-induced CXC chemokine (LIX), MIP-1α, MIP-1β, MIP-2, RANTES, nitric oxide, intracellular calcium, and hydrogen peroxide were 591.3, 450, 1719, 27.68, 369.4, 256.6, 230.7, 856.9, 1326, 1524, 378.1, 26.76, 345.1, and 32.95 μM, respectively. Baicalin modulated the inflammatory response of macrophages activated by LPS via the calcium-CHOP pathway.
Collapse
|
9
|
Alpha7 Nicotinic Acetylcholine Receptor Antagonists Prevent Meningitic Escherichia coli-Induced Blood–Brain Barrier Disruptions by Targeting the CISH/JAK2/STAT5b Axis. Biomedicines 2022; 10:biomedicines10102358. [PMID: 36289622 PMCID: PMC9598402 DOI: 10.3390/biomedicines10102358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
Despite the availability of antibiotics over the last several decades, excessive antibiotic treatments for bacterial sepsis and meningitis (BSM) in children may result in several adverse outcomes. Hematogenous pathogens may directly induce permeability increases in human brain microvascular endothelial cells (HBMECs) and blood–brain barrier (BBB) dysfunctions. Our preliminary studies demonstrated that the alpha7 nicotinic acetylcholine receptor (α7nAChR) played an important role in the pathogenesis of BSM, accompanied by increasing cytokine-inducible SH2-containing protein (CISH) at the transcriptome level, but it has remained unclear how α7nAChR-CISH works mechanistically. The study aims to explore the underlying mechanism of α7nAChR and CISH during E. coli-induced BSM in vitro (HBMECs) and in vivo (α7nAChR-KO mouse). We found that in the stage of E. coli K1-induced BBB disruptions, α7nAChR functioned as the key regulator that affects the integrity of HBMECs by activating the JAK2–STAT5 signaling pathway, while CISH inhibited JAK2–STAT5 activation and exhibited protective effects against E. coli infection. Notably, we first validated that the expression of CISH could be regulated by α7nAChR in HBMECs. In addition, we determined the protective effects of MLA (methyllycaconitine citrate) and MEM (memantine hydrochloride) (functioning as α7nAChR antagonists) on infected HBMECs and suggested that the α7nAChR–CISH axis could explain the protective effects of the two small-molecule compounds on E. coli-induced HBMECs injuries and BBB disruptions. In conclusion, we dissected the α7nAChR/CISH/JAK2/STAT5 axis as critical for the pathogenesis of E. coli-induced brain microvascular leakage and BBB disruptions and provided novel evidence for the development of α7nAChR antagonists in the prevention of pediatric E. coli BSM.
Collapse
|
10
|
Lipopolysaccharide-Induced Immunological Tolerance in Monocyte-Derived Dendritic Cells. IMMUNO 2022. [DOI: 10.3390/immuno2030030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Bacterial lipopolysaccharides (LPS), also referred to as endotoxins, are major outer surface membrane components present on almost all Gram-negative bacteria and are major determinants of sepsis-related clinical complications including septic shock. LPS acts as a strong stimulator of innate or natural immunity in a wide variety of eukaryotic species ranging from insects to humans including specific effects on the adaptive immune system. However, following immune stimulation, lipopolysaccharide can induce tolerance which is an essential immune-homeostatic response that prevents overactivation of the inflammatory response. The tolerance induced by LPS is a state of reduced immune responsiveness due to persistent and repeated challenges, resulting in decreased expression of pro-inflammatory modulators and up-regulation of antimicrobials and other mediators that promote a reduction of inflammation. The presence of environmental-derived LPS may play a key role in decreasing autoimmune diseases and gut tolerance to the plethora of ingested antigens. The use of LPS may be an important immune adjuvant as demonstrated by the promotion of IDO1 increase when present in the fusion protein complex of CTB-INS (a chimera of the cholera toxin B subunit linked to proinsulin) that inhibits human monocyte-derived DC (moDC) activation, which may act through an IDO1-dependent pathway. The resultant state of DC tolerance can be further enhanced by the presence of residual E. coli lipopolysaccharide (LPS) which is almost always present in partially purified CTB-INS preparations. The approach to using an adjuvant with an autoantigen in immunotherapy promises effective treatment for devastating tissue-specific autoimmune diseases like multiple sclerosis (MS) and type 1 diabetes (T1D).
Collapse
|
11
|
Sun JD, Li Q, Haoyang WW, Zhang DW, Wang H, Zhou W, Ma D, Hou JL, Li ZT. Adsorption-Based Detoxification of Endotoxins by Porous Flexible Organic Frameworks. Mol Pharm 2022; 19:953-962. [PMID: 35102736 DOI: 10.1021/acs.molpharmaceut.1c00923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Bacterial lipopolysaccharides (LPS, endotoxins) cause sepsis that is responsible for a huge amount of mortality globally. However, their neutralization or detoxification remains an unmet medical need. We envisaged that cationic organic frameworks with persistent hydrophobic porosity may adsorb and thus neutralize LPS through a combination of cooperative ion-pairing electrostatic attraction and hydrophobicity. We here report the preparation of two water-soluble flexible organic frameworks (FOF-1 and FOF-2) from tetratopic and ditopic precursors through quantitative formation of hydrazone bonds at room temperature. The two FOFs are revealed to possess hydrodynamic diameters, which range from 20 to 120 nm, depending on the concentrations. Dynamic light scattering and isothermal titration calorimetric and chromogenic limulus amebocyte lysate experiments indicate that both frameworks are able to adsorb and thus reduce the concentration of free LPS molecules in aqueous solution, whereas cytokine inhibition experiments with RAW264.7 support that this adsorption can significantly decrease the toxicity of LPS. In vivo experiments with mice (five males per group) show that the injection of FOF-1 at a dose of 0.6 mg/kg realizes the survival of all of the mice administrated with LPS of the d-galactosamine (d-Gal)-sensitized absolute lethal dose (LD100, 0.05 mg/kg), whereas its maximum tolerated dose for mice is determined to be 10 mg/kg. These findings provide a new promising sequestration strategy for the development of porous agents for the neutralization of LPS.
Collapse
Affiliation(s)
- Jian-Da Sun
- Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China
| | - Qian Li
- Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China
| | - Wei-Wei Haoyang
- Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China
| | - Dan-Wei Zhang
- Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China
| | - Hui Wang
- Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China
| | - Wei Zhou
- Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China
| | - Da Ma
- Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China
| | - Jun-Li Hou
- Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China
| | - Zhan-Ting Li
- Department of Chemistry, Fudan University, 2205 Songhu Road, Shanghai 200438, China.,Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, 2205 Songhu Road, Shanghai 200438, China
| |
Collapse
|
12
|
How to Combat Gram-Negative Bacteria Using Antimicrobial Peptides: A Challenge or an Unattainable Goal? Antibiotics (Basel) 2021; 10:antibiotics10121499. [PMID: 34943713 PMCID: PMC8698890 DOI: 10.3390/antibiotics10121499] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 12/16/2022] Open
Abstract
Antimicrobial peptides (AMPs) represent a promising and effective alternative for combating pathogens, having some advantages compared to conventional antibiotics. However, AMPs must also contend with complex and specialised Gram-negative bacteria envelops. The variety of lipopolysaccharide and phospholipid composition in Gram-negative bacteria strains and species are decisive characteristics regarding their susceptibility or resistance to AMPs. Such biological and structural barriers have created delays in tuning AMPs to deal with Gram-negative bacteria. This becomes even more acute because little is known about the interaction AMP–Gram-negative bacteria and/or AMPs’ physicochemical characteristics, which could lead to obtaining selective molecules against Gram-negative bacteria. As a consequence, available AMPs usually have highly associated haemolytic and/or cytotoxic activity. Only one AMP has so far been FDA approved and another two are currently in clinical trials against Gram-negative bacteria. Such a pessimistic panorama suggests that efforts should be concentrated on the search for new molecules, designs and strategies for combating infection caused by this type of microorganism. This review has therefore been aimed at describing the currently available AMPs for combating Gram-negative bacteria, exploring the characteristics of these bacteria’s cell envelop hampering the development of new AMPs, and offers a perspective regarding the challenges for designing new AMPs against Gram-negative bacteria.
Collapse
|
13
|
Fock EM, Parnova RG. Protective Effect of Mitochondria-Targeted Antioxidants against Inflammatory Response to Lipopolysaccharide Challenge: A Review. Pharmaceutics 2021; 13:pharmaceutics13020144. [PMID: 33499252 PMCID: PMC7910823 DOI: 10.3390/pharmaceutics13020144] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/15/2021] [Accepted: 01/17/2021] [Indexed: 12/16/2022] Open
Abstract
Lipopolysaccharide (LPS), the major component of the outer membrane of Gram-negative bacteria, is the most abundant proinflammatory agent. Considerable evidence indicates that LPS challenge inescapably causes oxidative stress and mitochondrial dysfunction, leading to cell and tissue damage. Increased mitochondrial reactive oxygen species (mtROS) generation triggered by LPS is known to play a key role in the progression of the inflammatory response. mtROS at excessive levels impair electron transport chain functioning, reduce the mitochondrial membrane potential, and initiate lipid peroxidation and oxidative damage of mitochondrial proteins and mtDNA. Over the past 20 years, a large number of mitochondria-targeted antioxidants (mito-AOX) of different structures that can accumulate inside mitochondria and scavenge free radicals have been synthesized. Their protective role based on the prevention of oxidative stress and the restoration of mitochondrial function has been demonstrated in a variety of common diseases and pathological states. This paper reviews the current data on the beneficial application of different mito-AOX in animal endotoxemia models, in either in vivo or in vitro experiments. The results presented in our review demonstrate the promising potential of approaches based on mito-AOX in the development of new treatment strategies against Gram-negative infections and LPS per se.
Collapse
|