1
|
Wang Z, Yang J, Zhang DE, Qiao X, Yang SL, Wang ZH, Yang Q. Doxycycline-Induced Apoptosis in Brucella suis S2-Infected HMC3 Cells via Calreticulin Suppression and Activation of the IRE1/Caspase-3 Signaling Pathway. Infect Drug Resist 2025; 18:2005-2020. [PMID: 40290405 PMCID: PMC12034290 DOI: 10.2147/idr.s507193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
Objective This study aims to elucidate the apoptotic mechanism induced by doxycycline (Dox) in human microglial clone 3 (HMC3) cells infected with the Brucella suis S2 strain, with the goal of identifying potential therapeutic targets for neurobrucellosis. Methods The expression of calreticulin (CALR) at both the protein and mRNA levels was assessed using Western blot analysis and reverse transcription-quantitative polymerase chain reaction (RT-qPCR), respectively, following exposure of HMC3 cells to varying concentrations and treatment durations of Dox. Apoptosis rates were determined via flow cytometry. To investigate the involvement of the inositol-requiring enzyme-1 (IRE1)/Caspase-12/Caspase-3 pathway, CALR protein levels were analyzed through Western blot after a 12-hour treatment with 160 μM Dox. Endoplasmic reticulum (ER) stress and intracellular calcium (Ca²⁺) concentrations were evaluated using fluorescent staining. The same parameters were measured in B. suis S2-infected HMC3 cells following treatment with 160 μM Dox. Results Treatment with 160 μM Dox for 12 hours resulted in a reduction in CALR protein levels and the induction of apoptosis in HMC3 cells. The downregulation of CALR activated the IRE1/Caspase-12/Caspase-3 signaling pathway, leading to apoptosis. Similar apoptotic effects were observed in B. suis S2-infected HMC3 cells following Dox treatment. Conclusion Dox promotes apoptosis in B. suis S2-infected HMC3 cells by suppressing CALR expression and activating the IRE1/Caspase-12/Caspase-3 signaling pathway. These findings suggest that CALR regulation may serve as a potential therapeutic target for neurobrucellosis.
Collapse
Affiliation(s)
- Zhao Wang
- Department of Experimental Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi’an, People’s Republic of China
| | - Juan Yang
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Deng-Er Zhang
- The First Clinical Medical School, Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Xia Qiao
- Institute of Medical Science, The General Hospital of Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Shu-Long Yang
- Department of Orthopedics, The People’s Hospital of Wuhai, Wuhai, People’s Republic of China
| | - Zhen-Hai Wang
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, People’s Republic of China
- Diagnosis and Treatment Engineering Technology Research Center of Nervous System Diseases of Ningxia Hui Autonomous Region, The General Hospital of Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Qian Yang
- Department of Experimental Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi’an, People’s Republic of China
| |
Collapse
|
2
|
Ma L, Liu Y, Sun J, Yang X, He Y, Zhang T, Zhao J, Lu Z, Yan X, Qie X. The synthesis of nitric oxide regulated by JNK pathway in the pea aphid to defend against bacterial infection. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 180:104315. [PMID: 40274239 DOI: 10.1016/j.ibmb.2025.104315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/13/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
Compared to other insects, the pea aphid Acyrthosiphon pisum exhibits limited immune responses, particularly due to the absence of many immune genes, including those encoding antimicrobial peptides and key components of the IMD pathway. Prior studies proved that the conserved signaling, Jun N-terminal kinase (JNK) pathway, plays a critical role in the immune system of the pea aphid, and nitric oxide synthase (NOS) is required for the pea aphid's defense against infections. Herein, using in vitro biochemical assays and in vivo bioassays, we demonstrated that the JNK pathway directly regulates the expression of NOS and that the JNK pathway-NOS-NO signal axis is efficient in defending against bacterial infections. The Toll pathway is instrumental for combating bacterial infections, and NO can activate the Toll pathway. The Toll pathway induced by NO regulates the expressions of ROS metabolism, lysosome, and phagocytosis-related genes. NO was identified as a crucial signaling molecule that facilitates communication between the JNK and Toll pathways.
Collapse
Affiliation(s)
- Li Ma
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Yaya Liu
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Jing Sun
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Xiaorong Yang
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Yingying He
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Tingting Zhang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Jingyu Zhao
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Zhiqiang Lu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Xizhong Yan
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China.
| | - Xingtao Qie
- College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China.
| |
Collapse
|
3
|
Awogbindin I, ŠimonČiČová E, Vidal V, Ash C, Tremblay ME. Neuroglial responses to bacterial, viral, and fungal neuroinfections. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:213-238. [PMID: 40148046 DOI: 10.1016/b978-0-443-19102-2.00027-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Evidence regarding the host's response to peripheral pathogens in humans abound, whereas studies on the pathogenesis of central nervous system-penetrating infections are relatively scarce. However, given the spate of epidemic and pandemic neuroinfections in the 21st century, the field has experienced a renewed interest lately. This chapter discusses a timely and exciting topic on the roles of glial cells, mainly microglia and astrocytes, in neuroinvasive infections. This chapter considered fungal, viral, and bacterial neuroinfections, X-raying their neuroinvasiveness, neurotropism, and neurovirulence before focusing on specific examples notable for each category, including Escherichia coli, Cryptococcus neoformans, and SARS-CoV-2. These infections are renowned worldwide for a high case-fatality rate, leaving many survivors with life-long morbidity and others with a bleak future neurologic prognosis. Importantly, the chapter discusses possible ways microglia and astrocytes are culpable in these infections and provides approaches by which they can be manipulated for therapeutic purposes, identifying viable research gaps in the process. Additionally, it offers a synopsis of ongoing works considering microglial selective targeting to attenuate the pathology, morbidity, and mortality associated with these neuroinfections. Considering that microglia and astrocytes are first responders in the central nervous system, targeting these glial cells could be the game changer in managing existing and emerging neuroinvasive infections.
Collapse
Affiliation(s)
- Ifeoluwa Awogbindin
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada
| | - Eva ŠimonČiČová
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Virginie Vidal
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Science and Technology Department, University of Bordeaux, Bordeaux, France
| | - Chantaille Ash
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Marie-Eve Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
4
|
Sodhi CP, Ahmad R, Fulton WB, Lopez CM, Eke BO, Scheese D, Duess JW, Steinway SN, Raouf Z, Moore H, Tsuboi K, Sampah ME, Jang HS, Buck RH, Hill DR, Niemiro GM, Prindle T, Wang S, Wang M, Jia H, Catazaro J, Lu P, Hackam DJ. Human milk oligosaccharides reduce necrotizing enterocolitis-induced neuroinflammation and cognitive impairment in mice. Am J Physiol Gastrointest Liver Physiol 2023; 325:G23-G41. [PMID: 37120853 PMCID: PMC10259852 DOI: 10.1152/ajpgi.00233.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/02/2023]
Abstract
Necrotizing enterocolitis (NEC) is the leading cause of morbidity and mortality in premature infants. One of the most devastating complications of NEC is the development of NEC-induced brain injury, which manifests as impaired cognition that persists beyond infancy and which represents a proinflammatory activation of the gut-brain axis. Given that oral administration of the human milk oligosaccharides (HMOs) 2'-fucosyllactose (2'-FL) and 6'-sialyslactose (6'-SL) significantly reduced intestinal inflammation in mice, we hypothesized that oral administration of these HMOs would reduce NEC-induced brain injury and sought to determine the mechanisms involved. We now show that the administration of either 2'-FL or 6'-SL significantly attenuated NEC-induced brain injury, reversed myelin loss in the corpus callosum and midbrain of newborn mice, and prevented the impaired cognition observed in mice with NEC-induced brain injury. In seeking to define the mechanisms involved, 2'-FL or 6'-SL administration resulted in a restoration of the blood-brain barrier in newborn mice and also had a direct anti-inflammatory effect on the brain as revealed through the study of brain organoids. Metabolites of 2'-FL were detected in the infant mouse brain by nuclear magnetic resonance (NMR), whereas intact 2'-FL was not. Strikingly, the beneficial effects of 2'-FL or 6'-SL against NEC-induced brain injury required the release of the neurotrophic factor brain-derived neurotrophic factor (BDNF), as mice lacking BDNF were not protected by these HMOs from the development of NEC-induced brain injury. Taken in aggregate, these findings reveal that the HMOs 2'-FL and 6'-SL interrupt the gut-brain inflammatory axis and reduce the risk of NEC-induced brain injury.NEW & NOTEWORTHY This study reveals that the administration of human milk oligosaccharides, which are present in human breast milk, can interfere with the proinflammatory gut-brain axis and prevent neuroinflammation in the setting of necrotizing enterocolitis, a major intestinal disorder seen in premature infants.
Collapse
Affiliation(s)
- Chhinder P Sodhi
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Raheel Ahmad
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - William B Fulton
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Carla M Lopez
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Benjamin O Eke
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Daniel Scheese
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Johannes W Duess
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Steve N Steinway
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Zachariah Raouf
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Hannah Moore
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Koichi Tsuboi
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Maame Efua Sampah
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Hee-Seong Jang
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Rachael H Buck
- Nutrition Division, Abbott, Columbus, Ohio, United States
| | - David R Hill
- Nutrition Division, Abbott, Columbus, Ohio, United States
| | | | - Thomas Prindle
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Sanxia Wang
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Menghan Wang
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Hongpeng Jia
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - Jonathan Catazaro
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland, United States
| | - Peng Lu
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| | - David J Hackam
- Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Department of Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Baltimore, Maryland, United States
| |
Collapse
|
5
|
The Pathological Activation of Microglia Is Modulated by Sexually Dimorphic Pathways. Int J Mol Sci 2023; 24:ijms24054739. [PMID: 36902168 PMCID: PMC10003784 DOI: 10.3390/ijms24054739] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/11/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Microglia are the primary immunocompetent cells of the central nervous system (CNS). Their ability to survey, assess and respond to perturbations in their local environment is critical in their role of maintaining CNS homeostasis in health and disease. Microglia also have the capability of functioning in a heterogeneous manner depending on the nature of their local cues, as they can become activated on a spectrum from pro-inflammatory neurotoxic responses to anti-inflammatory protective responses. This review seeks to define the developmental and environmental cues that support microglial polarization towards these phenotypes, as well as discuss sexually dimorphic factors that can influence this process. Further, we describe a variety of CNS disorders including autoimmune disease, infection, and cancer that demonstrate disparities in disease severity or diagnosis rates between males and females, and posit that microglial sexual dimorphism underlies these differences. Understanding the mechanism behind differential CNS disease outcomes between men and women is crucial in the development of more effective targeted therapies.
Collapse
|
6
|
Li S, Wernersbach I, Harms GS, Schäfer MKE. Microglia subtypes show substrate- and time-dependent phagocytosis preferences and phenotype plasticity. Front Immunol 2022. [PMID: 36105813 DOI: 10.3389/fimmu.2022b.945485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
Microglia are phagocytosis-competent CNS cells comprising a spectrum of subtypes with beneficial and/or detrimental functions in acute and chronic neurodegenerative disorders. The heterogeneity of microglia suggests differences in phagocytic activity and phenotype plasticity between microglia subtypes. To study these issues, primary murine glial cultures were cultivated in the presence of serum, different growth factors and cytokines to obtain M0-like, M1-like, and M2-like microglia as confirmed by morphology, M1/M2 gene marker expression, and nitric oxide assay. Single-cell analysis after 3 hours of phagocytosis of E.coli particles or IgG-opsonized beads showed equal internalization by M0-like microglia, whereas M1-like microglia preferably internalized E.coli particles and M2-like microglia preferably internalized IgG beads, suggesting subtype-specific preferences for different phagocytosis substrates. Time-lapse live-cells imaging over 16 hours revealed further differences between microglia subtypes in phagocytosis preference and internalization dynamics. M0- and, more efficiently, M1-like microglia continuously internalized E.coli particles for 16 hours, whereas M2-like microglia discontinued internalization after approximately 8 hours. IgG beads were continuously internalized by M0- and M1-like microglia but strikingly less by M2-like microglia. M2-like microglia initially showed continuous internalization similar to M0-like microglia but again discontinuation of internalization after 8 hours suggesting that the time of substrate exposure differently affect microglia subtypes. After prolonged exposure to E.coli particles or IgG beads for 5 days all microglia subtypes showed increased internalization of E.coli particles compared to IgG beads, increased nitric oxide release and up-regulation of M1 gene markers, irrespectively of the phagocytosis substrate, suggesting phenotype plasticity. In summary, microglia subtypes show substrate- and time-dependent phagocytosis preferences and phenotype plasticity. The results suggest that prolonged phagocytosis substrate exposure enhances M1-like profiles and M2-M1 repolarization of microglia. Similar processes may also take place in conditions of acute and chronic brain insults when microglia encounter different types of phagocytic substrates.
Collapse
Affiliation(s)
- Shuailong Li
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Isa Wernersbach
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Gregory S Harms
- Cell Biology Unit, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany.,Departments of Biology and Physics, Wilkes University, Wilkes Barre, PA, United States
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany.,Focus Program Translational Neurosciences (FTN), Johannes Gutenberg-University Mainz, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
7
|
Li S, Wernersbach I, Harms GS, Schäfer MKE. Microglia subtypes show substrate- and time-dependent phagocytosis preferences and phenotype plasticity. Front Immunol 2022; 13:945485. [PMID: 36105813 PMCID: PMC9465456 DOI: 10.3389/fimmu.2022.945485] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Microglia are phagocytosis-competent CNS cells comprising a spectrum of subtypes with beneficial and/or detrimental functions in acute and chronic neurodegenerative disorders. The heterogeneity of microglia suggests differences in phagocytic activity and phenotype plasticity between microglia subtypes. To study these issues, primary murine glial cultures were cultivated in the presence of serum, different growth factors and cytokines to obtain M0-like, M1-like, and M2-like microglia as confirmed by morphology, M1/M2 gene marker expression, and nitric oxide assay. Single-cell analysis after 3 hours of phagocytosis of E.coli particles or IgG-opsonized beads showed equal internalization by M0-like microglia, whereas M1-like microglia preferably internalized E.coli particles and M2-like microglia preferably internalized IgG beads, suggesting subtype-specific preferences for different phagocytosis substrates. Time-lapse live-cells imaging over 16 hours revealed further differences between microglia subtypes in phagocytosis preference and internalization dynamics. M0- and, more efficiently, M1-like microglia continuously internalized E.coli particles for 16 hours, whereas M2-like microglia discontinued internalization after approximately 8 hours. IgG beads were continuously internalized by M0- and M1-like microglia but strikingly less by M2-like microglia. M2-like microglia initially showed continuous internalization similar to M0-like microglia but again discontinuation of internalization after 8 hours suggesting that the time of substrate exposure differently affect microglia subtypes. After prolonged exposure to E.coli particles or IgG beads for 5 days all microglia subtypes showed increased internalization of E.coli particles compared to IgG beads, increased nitric oxide release and up-regulation of M1 gene markers, irrespectively of the phagocytosis substrate, suggesting phenotype plasticity. In summary, microglia subtypes show substrate- and time-dependent phagocytosis preferences and phenotype plasticity. The results suggest that prolonged phagocytosis substrate exposure enhances M1-like profiles and M2-M1 repolarization of microglia. Similar processes may also take place in conditions of acute and chronic brain insults when microglia encounter different types of phagocytic substrates.
Collapse
Affiliation(s)
- Shuailong Li
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Isa Wernersbach
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Gregory S. Harms
- Cell Biology Unit, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
- Departments of Biology and Physics, Wilkes University, Wilkes Barre, PA, United States
| | - Michael K. E. Schäfer
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
- Focus Program Translational Neurosciences (FTN), Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
- *Correspondence: Michael K. E. Schäfer,
| |
Collapse
|
8
|
Synthetic TGF-β Signaling Agonist-Treated Dendritic Cells Induce Tolerogenicity and Antirheumatic Effects. Curr Issues Mol Biol 2022; 44:3809-3821. [PMID: 36135173 PMCID: PMC9498161 DOI: 10.3390/cimb44090261] [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: 06/10/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
The newly synthesized compound TGF-β signaling agonist (T74) is a small molecule associated with the TGF-β receptor signaling pathway. Tolerogenic dendritic cells (tDCs) have been used to examine immunosuppressive and anti-inflammatory effects in multiple autoimmune disease models. The aim of this study was to investigate whether treatment of DCs with T74 has an antirheumatic effect in a mouse model of collagen-induced arthritis (CIA). Bone marrow-derived cells were obtained from DBA/1J mice and differentiated into DCs. T74-treated DCs (T74-DCs) were generated by treating bone marrow-derived DCs with LPS, type II collagen, and T74. T74-DCs expressed lower levels of surface molecules and inflammatory cytokines associated with antigen presentation and T cell stimulation. The ability of T74-DCs to differentiate effector T cells was lower than that of T74-untreated DCs (NT-DCs), but T74-DCs increased the regulatory T (Treg) cell differentiation in vitro. DBA/1J mice received two subcutaneous (s.c.) injections of type II collagen to establish CIA. Mice then received two s.c. injections of T74-DCs or NT-DCs. Joint inflammation was ameliorated in the paws of T74-DC-treated mice. Additionally, Treg populations in T74-DC-treated mice were higher than in NT-DC-treated or PBS-treated CIA mice. Taken together, these results demonstrate that T74 induces tolerance in DCs, and that T74-mediated DCs exert antirheumatic effects via induction of Tregs.
Collapse
|
9
|
Galectin-3, a rising star in modulating microglia activation under conditions of neurodegeneration. Cell Death Dis 2022; 13:628. [PMID: 35859075 PMCID: PMC9300700 DOI: 10.1038/s41419-022-05058-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 01/21/2023]
Abstract
The advent of high-throughput single-cell transcriptomic analysis of microglia has revealed different phenotypes that are inherently associated with disease conditions. A common feature of some of these activated phenotypes is the upregulation of galectin-3. Representative examples of these phenotypes include disease-associated microglia (DAM) and white-associated microglia (WAM), whose role(s) in neuroprotection/neurotoxicity is a matter of high interest in the microglia community. In this review, we summarise the main findings that demonstrate the ability of galectin-3 to interact with key pattern recognition receptors, including, among others, TLR4 and TREM2 and the importance of galectin-3 in the regulation of microglia activation. Finally, we discuss increasing evidence supporting the involvement of this lectin in the main neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, traumatic brain injury, and stroke.
Collapse
|
10
|
Bacteria reduce flagellin synthesis to evade microglia-astrocyte-driven immunity in the brain. Cell Rep 2022; 40:111033. [PMID: 35793624 DOI: 10.1016/j.celrep.2022.111033] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 05/09/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
The immune response of brain cells to invading bacteria in vivo and the mechanism used by pathogenic bacteria to escape brain immune surveillance remain largely unknown. It is believed that microglia eliminate bacteria by phagocytosis based on in vitro data. Here we find that a small percentage of microglia in the brain engulf neonatal meningitis-causing Escherichia coli (NMEC), but more microglia are activated to produce tumor necrosis factor alpha (TNFα), which activates astrocytes to secrete complement component 3 (C3) involved in anti-bacterial activity. To evade anti-bacterial activity of the immune system, NMEC senses low concentration of threonine in cerebrospinal fluid (CSF) to down-modulate the expression of flagellin and reduce microglial TNFα and astrocyte C3 production. Our findings may help develop strategies for bacterial meningitis treatment.
Collapse
|
11
|
Activin A is a novel chemoattractant for migration of microglial BV2 cells. J Neuroimmunol 2022; 371:577929. [DOI: 10.1016/j.jneuroim.2022.577929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/08/2022] [Accepted: 07/09/2022] [Indexed: 11/19/2022]
|
12
|
Liu P, Wang X, Yang Q, Yan X, Fan Y, Zhang S, Wei Y, Huang M, Jiang L, Feng L. Collaborative Action of Microglia and Astrocytes Mediates Neutrophil Recruitment to the CNS to Defend against Escherichia coli K1 Infection. Int J Mol Sci 2022; 23:ijms23126540. [PMID: 35742984 PMCID: PMC9223767 DOI: 10.3390/ijms23126540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 02/04/2023] Open
Abstract
Escherichia coli K1 is a leading cause of neonatal bacterial meningitis. Recruitment of neutrophils to the central nervous system (CNS) via local immune response plays a critical role in defense against E. coli K1 infection; however, the mechanism underlying this recruitment remains unclear. In this study, we report that microglia and astrocytes are activated in response to stimulation by E. coli K1 and/or E. coli K1-derived outer membrane vesicles (OMVs) and work collaboratively to drive neutrophil recruitment to the CNS. Microglial activation results in the release of the pro-inflammatory cytokine TNF-α, which activates astrocytes, resulting in the production of CXCL1, a chemokine critical for recruiting neutrophils. Mice lacking either microglia or TNF-α exhibit impaired production of CXCL1, impaired neutrophil recruitment, and an increased CNS bacterial burden. C-X-C chemokine receptor 2 (CXCR2)-expressing neutrophils primarily respond to CXCL1 released by astrocytes. This study provides further insights into how immune responses drive neutrophil recruitment to the brain to combat E. coli K1 infection. In addition, we show that direct recognition of E. coli K1 by microglia is prevented by the K1 capsule. This study also reveals that OMVs are sufficient to induce microglial activation.
Collapse
Affiliation(s)
- Peng Liu
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300457, China; (P.L.); (X.W.); (Q.Y.); (X.Y.); (Y.F.); (S.Z.); (Y.W.); (M.H.)
- Tianjin Key Laboratory of Microbial Functional Genomics, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China
| | - Xinyue Wang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300457, China; (P.L.); (X.W.); (Q.Y.); (X.Y.); (Y.F.); (S.Z.); (Y.W.); (M.H.)
- Tianjin Key Laboratory of Microbial Functional Genomics, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China
| | - Qian Yang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300457, China; (P.L.); (X.W.); (Q.Y.); (X.Y.); (Y.F.); (S.Z.); (Y.W.); (M.H.)
- Tianjin Key Laboratory of Microbial Functional Genomics, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China
| | - Xiaolin Yan
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300457, China; (P.L.); (X.W.); (Q.Y.); (X.Y.); (Y.F.); (S.Z.); (Y.W.); (M.H.)
- Tianjin Key Laboratory of Microbial Functional Genomics, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China
| | - Yu Fan
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300457, China; (P.L.); (X.W.); (Q.Y.); (X.Y.); (Y.F.); (S.Z.); (Y.W.); (M.H.)
- Tianjin Key Laboratory of Microbial Functional Genomics, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China
| | - Si Zhang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300457, China; (P.L.); (X.W.); (Q.Y.); (X.Y.); (Y.F.); (S.Z.); (Y.W.); (M.H.)
- Tianjin Key Laboratory of Microbial Functional Genomics, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China
| | - Yi Wei
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300457, China; (P.L.); (X.W.); (Q.Y.); (X.Y.); (Y.F.); (S.Z.); (Y.W.); (M.H.)
- Tianjin Key Laboratory of Microbial Functional Genomics, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China
| | - Min Huang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300457, China; (P.L.); (X.W.); (Q.Y.); (X.Y.); (Y.F.); (S.Z.); (Y.W.); (M.H.)
- Tianjin Key Laboratory of Microbial Functional Genomics, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China
| | - Lingyan Jiang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300457, China; (P.L.); (X.W.); (Q.Y.); (X.Y.); (Y.F.); (S.Z.); (Y.W.); (M.H.)
- Tianjin Key Laboratory of Microbial Functional Genomics, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China
- Correspondence: (L.J.); (L.F.)
| | - Lu Feng
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin 300457, China; (P.L.); (X.W.); (Q.Y.); (X.Y.); (Y.F.); (S.Z.); (Y.W.); (M.H.)
- Tianjin Key Laboratory of Microbial Functional Genomics, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China
- Correspondence: (L.J.); (L.F.)
| |
Collapse
|
13
|
Fei ZY, Wang J, Liang J, Zhou X, Guo M. Analysis of bacterial spectrum, activin A, and CD64 in chronic obstructive pulmonary disease patients complicated with pulmonary infections. World J Clin Cases 2022; 10:2382-2392. [PMID: 35434072 PMCID: PMC8968607 DOI: 10.12998/wjcc.v10.i8.2382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/27/2021] [Accepted: 01/29/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pulmonary infections often lead to poor prognoses in patients with chronic obstructive pulmonary disease (COPD). Activin A and CD64 play crucial pathological roles in the development of COPD.
AIM To explore the bacterial spectrum via analysis of activing A levels, CD64 index, and related mechanisms in COPD patients complicated with pulmonary infection.
METHODS Between March 2015 and January 2018, a total of 85 patients with COPD, who also suffered from pulmonary infections, were enrolled in this study as the pulmonary infection group. In addition, a total of 96 COPD patients, without pulmonary infection, were selected as the control group. Sputum samples of patients in the pulmonary infection group were cultivated for bacterial identification prior to administration of antibiotics. The neutrophil CD64 index was measured using flow cytometry, serum activin A levels were detected via an enzyme-linked immunosorbent assay, and activin A, Smad3, TLR4, MyD88, and NFκB protein expression was analyzed by Western blotting.
RESULTS Gram-negative bacteria were identified in 57.65% of the sputum samples in the pulmonary infection group. The most prevalent Gram-negative species were Pseudomonas aeruginosa and Klebsiella pneumoniae. Conversely, Gram-positive bacteria were identified in 41.18% of the sputum samples in the pulmonary infection group. The most common Gram-positive species was Streptococcus pneumoniae. Fungi were identified in 1.17% of the sputum samples in the pulmonary infection group. The CD64 index was significantly higher in the pulmonary infection group (0.91 ± 0.38) than in the control group (0.23 ± 0.14, P < 0.001). The serum activin A levels were significantly higher in the pulmonary infection group (43.50 ± 5.22 ng/mL), compared to the control group (34.82 ± 4.16 ng/mL, P < 0.001). The relative expression levels of activin A, Smad3, TLR4, MyD88, and NFκB were all significantly higher in the pulmonary infection group, compared to the control group (all P < 0.001).
CONCLUSION Pulmonary infections in COPD patients are mainly caused by Streptococcus pneumoniae, Pseudomonas aeruginosa, and Klebsiella pneumoniae. Pulmonary infections can significantly increase neutrophil CD64 index and serum levels of activin A, thereby activating the activin A/Smad3 signaling pathway, which may positively regulate the TLR4/MyD88/NFκB signaling pathway.
Collapse
Affiliation(s)
- Zhao-Yang Fei
- Experimental Research Centre, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jiang Wang
- Experimental Research Centre, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jie Liang
- Experimental Research Centre, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xue Zhou
- Experimental Research Centre, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Min Guo
- Department of Laboratory Medicine, Lianyungang Second People's Hospital, Lianyungang 222006, Jiangsu Province, China
| |
Collapse
|
14
|
Zhang D, Xu S, Wang Y, Zhu G. The Potentials of Melatonin in the Prevention and Treatment of Bacterial Meningitis Disease. Molecules 2021; 26:1419. [PMID: 33808027 PMCID: PMC7961363 DOI: 10.3390/molecules26051419] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/23/2021] [Accepted: 03/02/2021] [Indexed: 02/08/2023] Open
Abstract
Bacterial meningitis (BM) is an acute infectious central nervous system (CNS) disease worldwide, occurring with 50% of the survivors left with a long-term serious sequela. Acute bacterial meningitis is more prevalent in resource-poor than resource-rich areas. The pathogenesis of BM involves complex mechanisms that are related to bacterial survival and multiplication in the bloodstream, increased permeability of blood-brain barrier (BBB), oxidative stress, and excessive inflammatory response in CNS. Considering drug-resistant bacteria increases the difficulty of meningitis treatment and the vaccine also has been limited to several serotypes, and the morbidity rate of BM still is very high. With recent development in neurology, there is promising progress for drug supplements of effectively preventing and treating BM. Several in vivo and in vitro studies have elaborated on understanding the significant mechanism of melatonin on BM. Melatonin is mainly secreted in the pineal gland and can cross the BBB. Melatonin and its metabolite have been reported as effective antioxidants and anti-inflammation, which are potentially useful as prevention and treatment therapy of BM. In bacterial meningitis, melatonin can play multiple protection effects in BM through various mechanisms, including immune response, antibacterial ability, the protection of BBB integrity, free radical scavenging, anti-inflammation, signaling pathways, and gut microbiome. This manuscript summarizes the major neuroprotective mechanisms of melatonin and explores the potential prevention and treatment approaches aimed at reducing morbidity and alleviating nerve injury of BM.
Collapse
Affiliation(s)
- Dong Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (D.Z.); (S.X.); (Y.W.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Shu Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (D.Z.); (S.X.); (Y.W.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Yiting Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (D.Z.); (S.X.); (Y.W.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (D.Z.); (S.X.); (Y.W.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| |
Collapse
|
15
|
Bukrinsky MI, Mukhamedova N, Sviridov D. Lipid rafts and pathogens: the art of deception and exploitation. J Lipid Res 2020; 61:601-610. [PMID: 31615838 PMCID: PMC7193957 DOI: 10.1194/jlr.tr119000391] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/07/2019] [Indexed: 02/06/2023] Open
Abstract
Lipid rafts, solid regions of the plasma membrane enriched in cholesterol and glycosphingolipids, are essential parts of a cell. Functionally, lipid rafts present a platform that facilitates interaction of cells with the outside world. However, the unique properties of lipid rafts required to fulfill this function at the same time make them susceptible to exploitation by pathogens. Many steps of pathogen interaction with host cells, and sometimes all steps within the entire lifecycle of various pathogens, rely on host lipid rafts. Such steps as binding of pathogens to the host cells, invasion of intracellular parasites into the cell, the intracellular dwelling of parasites, microbial assembly and exit from the host cell, and microbe transfer from one cell to another all involve lipid rafts. Interaction also includes modification of lipid rafts in host cells, inflicted by pathogens from both inside and outside the cell, through contact or remotely, to advance pathogen replication, to utilize cellular resources, and/or to mitigate immune response. Here, we provide a systematic overview of how and why pathogens interact with and exploit host lipid rafts, as well as the consequences of this interaction for the host, locally and systemically, and for the microbe. We also raise the possibility of modulation of lipid rafts as a therapeutic approach against a variety of infectious agents.
Collapse
Affiliation(s)
- Michael I Bukrinsky
- Department of Microbiology, Immunology, and Tropical Medicine,George Washington University School of Medicine and Health Science, Washington, DC 20037
| | | | - Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne 3004, Australia. mailto:
| |
Collapse
|
16
|
Effects of Pseudomonas aeruginosa on Microglial-Derived Extracellular Vesicle Biogenesis and Composition. Pathogens 2019; 8:pathogens8040297. [PMID: 31847332 PMCID: PMC6963293 DOI: 10.3390/pathogens8040297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/29/2019] [Accepted: 12/10/2019] [Indexed: 12/17/2022] Open
Abstract
The packaging of molecular constituents inside extracellular vesicles (EVs) allows them to participate in intercellular communication and the transfer of biological molecules, however the role of EVs during bacterial infection is poorly understood. The goal of this study was to examine the effects of Pseudomonas aeruginosa (P. aeruginosa) infection on the biogenesis and composition of EVs derived from the mouse microglia cell line, BV-2. BV-2 cells were cultured in exosome-free media and infected with 0, 1.3 × 104, or 2.6 × 104 colony forming units per milliliter P. aeruginosa for 72 h. The results indicated that compared with the control group, BV-2 cell viability significantly decreased after P. aeruginosa infection and BV-2-derived EVs concentration decreased significantly in the P. aeruginosa-infected group. P. aeruginosa infection significantly decreased chemokine ligand 4 messenger RNA in BV-2-derived infected EVs, compared with the control group (p ≤ 0.05). This study also revealed that heat shock protein 70 (p ≤ 0.05) and heat shock protein 90β (p ≤ 0.001) levels of expression within EVs increased after P. aeruginosa infection. EV treatment with EVs derived from P. aeruginosa infection reduced cell viability of BV-2 cells. P. aeruginosa infection alters the expression of specific proteins and mRNA in EVs. Our study suggests that P. aeruginosa infection modulates EV biogenesis and composition, which may influence bacterial pathogenesis and infection.
Collapse
|
17
|
Kietzman C, Tuomanen E. Acute Bacterial Meningitis: Challenges to Better Antibiotic Therapy. ACS Infect Dis 2019; 5:1987-1995. [PMID: 31268283 DOI: 10.1021/acsinfecdis.9b00122] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bacterial meningitis is a medical emergency requiring highly bactericidal antibiotics to achieve cure. Many challenges exist to achieving optimal patient outcome. First, antibiotics must pass the blood brain barrier. Once in the subarachnoid space, achieving bactericidal therapy involves circumventing antibiotic resistance and, more commonly, antibiotic tolerance arising from the slow growth of bacteria in the nutrient poor cerebrospinal fluid. Finally, bactericidal therapy is most often bacteriolytic, and debris from lysis is highly inflammatory. Controlling damage from lytic products may require adjunctive therapy to prevent neuronal death. These challenges are an extreme example of the different requirements for treating infections in different body sites.
Collapse
Affiliation(s)
- Colin Kietzman
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Elaine Tuomanen
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| |
Collapse
|
18
|
Activin-A in the regulation of immunity in health and disease. J Autoimmun 2019; 104:102314. [PMID: 31416681 DOI: 10.1016/j.jaut.2019.102314] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 07/28/2019] [Indexed: 02/08/2023]
Abstract
The TGF-β superfamily of cytokines plays pivotal roles in the regulation of immune responses protecting against or contributing to diseases, such as, allergy, autoimmunity and cancer. Activin-A, a member of the TGF-β superfamily, was initially identified as an inducer of follicle-stimulating hormone secretion. Extensive research over the past decades illuminated fundamental roles for activin-A in essential biologic processes, including embryonic development, stem cell maintenance and differentiation, haematopoiesis, cell proliferation and tissue fibrosis. Activin-A signals through two type I and two type II receptors which, upon ligand binding, activate their kinase activity, phosphorylate the SMAD2 and 3 intracellular signaling mediators that form a complex with SMAD4, translocate to the nucleus and activate or silence gene expression. Most immune cell types, including macrophages, dendritic cells (DCs), T and B lymphocytes and natural killer cells have the capacity to produce and respond to activin-A, although not in a similar manner. In innate immune cells, including macrophages, DCs and neutrophils, activin-A exerts a broad range of pro- or anti-inflammatory functions depending on the cell maturation and activation status and the spatiotemporal context. Activin-A also controls the differentiation and effector functions of Th cell subsets, including Th9 cells, TFH cells, Tr1 Treg cells and Foxp3+ Treg cells. Moreover, activin-A affects B cell responses, enhancing mucosal IgA secretion and inhibiting pathogenic autoantibody production. Interestingly, an array of preclinical and clinical studies has highlighted crucial functions of activin-A in the initiation, propagation and resolution of human diseases, including autoimmune diseases, such as, systemic lupus erythematosus, rheumatoid arthritis and pulmonary alveolar proteinosis, in allergic disorders, including allergic asthma and atopic dermatitis, in cancer and in microbial infections. Here, we provide an overview of the biology of activin-A and its signaling pathways, summarize recent studies pertinent to the role of activin-A in the modulation of inflammation and immunity, and discuss the potential of targeting activin-A as a novel therapeutic approach for the control of inflammatory diseases.
Collapse
|
19
|
Thorsdottir S, Henriques-Normark B, Iovino F. The Role of Microglia in Bacterial Meningitis: Inflammatory Response, Experimental Models and New Neuroprotective Therapeutic Strategies. Front Microbiol 2019; 10:576. [PMID: 30967852 PMCID: PMC6442515 DOI: 10.3389/fmicb.2019.00576] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/06/2019] [Indexed: 12/11/2022] Open
Abstract
Microglia have a pivotal role in the pathophysiology of bacterial meningitis. The goal of this review is to provide an overview on how microglia respond to bacterial pathogens targeting the brain, how the interplay between microglia and bacteria can be studied experimentally, and possible ways to use gained knowledge to identify novel preventive and therapeutic strategies. We discuss the dual role of microglia in disease development, the beneficial functions crucial for bacterial clearing, and the destructive properties through triggering neuroinflammation, characterized by cytokine and chemokine release which leads to leukocyte trafficking through the brain vascular endothelium and breakdown of the blood-brain barrier integrity. Due to intrinsic complexity of microglia and up until recently lack of specific markers, the study of microglial response to bacterial pathogens is challenging. New experimental models and techniques open up possibilities to accelerate progress in the field. We review existing models and discuss possibilities and limitations. Finally, we summarize recent findings where bacterial virulence factors are identified to be important for the microglial response, and how manipulation of evoked responses could be used for therapeutic or preventive purposes. Among promising approaches are: modulations of microglia phenotype switching toward anti-inflammatory and phagocytic functions, the use of non-bacterolytic antimicrobials, preventing release of bacterial components into the neural milieu and consequential amplification of immune activation, and protection of the blood-brain barrier integrity.
Collapse
Affiliation(s)
- Sigrun Thorsdottir
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Bioclinicum, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| | - Birgitta Henriques-Normark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Bioclinicum, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden.,Singapore Centre for Environmental Life Sciences Engineering (SCELSE) and Lee Kong Chian School of Medicine (LKC), Nanyang Technological University (NTU), Singapore, Singapore
| | - Federico Iovino
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Bioclinicum, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
20
|
Zang L, Wang J, Ren Y, Liu W, Yu Y, Zhao S, Otkur W, Zhao Y, Hayashi T, Tashiro SI, Onodera S, Ikejima T. Activated toll-like receptor 4 is involved in oridonin-induced phagocytosis via promotion of migration and autophagy-lysosome pathway in RAW264.7 macrophages. Int Immunopharmacol 2019; 66:99-108. [DOI: 10.1016/j.intimp.2018.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/09/2018] [Accepted: 11/09/2018] [Indexed: 12/19/2022]
|