1
|
Huang G, Yang S, Long T, Gao Y, Lin G. Proteomic analysis of brain tissue from ducks with meningitis caused by Riemerella anatipestifer infection. Poult Sci 2024; 103:104059. [PMID: 39068696 PMCID: PMC11338091 DOI: 10.1016/j.psj.2024.104059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Riemerella anatipestifer is a Gram-negative, rod-shaped bacterium that is flagellated, non-budded, and encapsulated, measuring approximately 0.4 μm × 0.7 μm. After infecting ducklings with R. anatipestifer, the hosts exhibited pathological changes, such as bacterial meningitis, fibrinous pericarditis, and fibrinous peripheral hepatitis. The pathogenesis of meningitis caused by R. anatipestifer has not yet been elucidated. To investigate the key molecules or proteins involved in R. anatipestifer's penetration of the blood-brain barrier (BBB) and the subsequent development of duck meningitis, a duck meningitis model was established and characterized. Duckling brain tissues were collected and analyzed using 4D label-free proteomic technology. Differentially expressed proteins were analyzed using a series of bioinformatics methods and verified using RT-qPCR and Western-Blot. The results showed that the differentially expressed proteins were primarily related to intracellular transport, transport protein activity, and transmembrane transport protein activity, and were mainly enriched in pathways associated with reducing intercellular connections and adhesion and increasing cell migration and apoptosis. Thus, it is suggested that R. anatipestifer may penetrate the BBB via transcellular and paracellular pathways, causing neurological diseases such as meningitis. This study is the first to analyze R. anatipestifer-infected duckling brain tissue using proteomics, thus providing a direction for further research into the mechanisms of R. anatipestifer's penetration of the BBB.
Collapse
Affiliation(s)
- Guoliang Huang
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China
| | - Shengmei Yang
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China
| | - Ting Long
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China
| | - Yuhan Gao
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China
| | - Guozhen Lin
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China.
| |
Collapse
|
2
|
Tong H, Wang L, Zhang K, Shi J, Wu Y, Bao Y, Wang C. S100A6 Activates Kupffer Cells via the p-P38 and p-JNK Pathways to Induce Inflammation, Mononuclear/macrophage Infiltration Sterile Liver Injury in Mice. Inflammation 2023; 46:534-554. [PMID: 36484925 DOI: 10.1007/s10753-022-01750-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/17/2022] [Accepted: 09/30/2022] [Indexed: 12/13/2022]
Abstract
Noninfectious liver injury, including the effects of chemical material, drugs and diet, is a major cause of liver diseases worldwide. In chemical and drugs-induced liver injury, innate inflammatory responses are mediated by extracellular danger signals. The S100 protein can act as danger signals, which can promote the migration and chemotaxis of immune cells, promote the release of various inflammatory cytokines, and regulate the body's inflammatory and immune responses. However, the role of S100A6 in inflammatory response in chemical and drugs-induced sterile liver injury remains unclear. We constructed the model of sterile liver injury induced by carbon tetrachloride (CCl4)/Paracetamol (APAP) and performed RNA sequencing (RNA-seq) on the liver tissues after injury (days 2 and 5). We analyzed inflammatory protein secretion in the liver tissue supernatant by enzyme-linked immunosorbent assay (ELISA), determined the inflammation response by bioinformatic analysis during sterile liver injury, and assessed mononuclear/macrophage infiltration by immunohistochemistry and flow cytometry. Immunohistochemistry was used to analyze the location of S100A6. We conducted inflammatory factor expression analysis and molecular mechanistic studies in Kupffer cells (KCs) induced by S100A6 using quantitative reverse transcription-polymerase chain reaction (qRT-PCR), ELISA, and western blot in vitro experiments. We performed chemokine CCL2 expression analysis and molecular mechanism studies using the same method. We used a Transwell assay to show the infiltration of mononuclear/macrophage. We here observed that aggravated inflammatory response was shown in CCl4 and APAP-administrated mice, as evidenced by enhanced production of inflammatory cytokines (TNF-α, IL-1β), and elevated mononuclear/macrophage infiltration and activation of immunity. The expression of S100A6 was significantly increased on day 2 after sterile liver injury, which is primarily produced by injured liver cells. Mechanistic studies established that S100A6 activates Kupffer cells (KCs) via the p-P38, p-JNK and P65 pathways to induce inflammation in vitro. Furthermore, TNF-α can stimulate liver cells via the p-P38 and p-JNK pathways to produce CCL2 and promote the infiltration of mononuclear/macrophage. In summary, we showed that S100A6 plays an important role in regulating inflammation, thus influencing sterile liver injury. Our findings provide novel evidence that S100A6 can as a danger signal that contributes to pro-inflammatory activation through p-P38 and p-JNK pathways in CCl4 and APAP-induced sterile liver injury in mice. In addition, the inflammatory factor TNF-α induces a large amount of CCL2 production in normal liver cells surrounding the injured area through a paracrine action, which is chemotactic for blood mononuclear/macrophage infiltration.
Collapse
Affiliation(s)
- He Tong
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Li Wang
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, Inner Mongolia, China
| | - Kefan Zhang
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Jing Shi
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Yongshuai Wu
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Yulong Bao
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, Inner Mongolia, China.
| | - Changshan Wang
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China.
| |
Collapse
|
3
|
Yang F, Cabe M, Nowak HA, Langert KA. Chitosan/poly(lactic-co-glycolic)acid Nanoparticle Formulations with Finely-Tuned Size Distributions for Enhanced Mucoadhesion. Pharmaceutics 2022; 14:95. [PMID: 35056991 PMCID: PMC8778482 DOI: 10.3390/pharmaceutics14010095] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/21/2021] [Accepted: 12/28/2021] [Indexed: 02/04/2023] Open
Abstract
Non-parenteral drug delivery systems using biomaterials have advantages over traditional parenteral strategies. For ocular and intranasal delivery, nanoparticulate systems must bind to and permeate through mucosal epithelium and other biological barriers. The incorporation of mucoadhesive and permeation-enhancing biomaterials such as chitosan facilitate this, but tend to increase the size and polydispersity of the nanoparticles, making practical optimization and implementation of mucoadhesive nanoparticle formulations a challenge. In this study, we adjusted key poly(lactic-co-glycolic) acid (PLGA) nanoparticle formulation parameters including the organic solvent and co-solvent, the concentration of polymer in the organic phase, the composition of the aqueous phase, the sonication amplitude, and the inclusion of chitosan in the aqueous phase. By doing so, we prepared four statistically unique size groups of PLGA NPs and equally-sized chitosan-PLGA NP counterparts. We loaded simvastatin, a candidate for novel ocular and intranasal delivery systems, into the nanoparticles to investigate the effects of size and surface modification on drug loading and release, and we quantified size- and surface-dependent changes in mucoadhesion in vitro. These methods and findings will contribute to the advancement of mucoadhesive nanoformulations for ocular and nose-to-brain drug delivery.
Collapse
Affiliation(s)
- Feipeng Yang
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, USA
- Research Service, Edward Hines, Jr., VA Hospital, Hines, IL 60141, USA
| | - Maleen Cabe
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, USA
- Research Service, Edward Hines, Jr., VA Hospital, Hines, IL 60141, USA
| | - Hope A Nowak
- Research Service, Edward Hines, Jr., VA Hospital, Hines, IL 60141, USA
| | - Kelly A Langert
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, USA
- Research Service, Edward Hines, Jr., VA Hospital, Hines, IL 60141, USA
| |
Collapse
|
4
|
Radaei Z, Zamani A, Najafi R, Saidijam M, Jalilian FA, Ezati R, Solgi G, Amini R. Promising Effects of Zerumbone on the Regulation of Tumor-promoting Cytokines Induced by TNF-α-activated Fibroblasts. Curr Med Sci 2021; 40:1075-1084. [PMID: 33428135 DOI: 10.1007/s11596-020-2289-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/26/2019] [Indexed: 11/28/2022]
Abstract
Inflammation plays an important role in the development of several cancers. Inflammatory cytokines, including tumor necrosis factor-α (TNF-α), are associated with the induction of inflammation. Chronic inflammation contributes to the progression of cancer through several mechanisms, including increased cytokine production and activation of transcription factors, such as nuclear factor-κB (NF-κB). Zerumbone (ZER), a component of subtropical ginger (Zingiber zerumbet Smith), seems to have anti-inflammatory, anti-cancer, and antioxidant activities. In this study, we aimed to explore the protective function and mechanisms of ZER against TNF-α-induced cancer-promoting cytokines. We found that the viability of stimulated human fibroblast cell lines was reduced after treatment with ZER (IC50=18 µmol/L), compared to un-stimulated fibroblasts (IC50=40 µmol/L). Besides, ZER inhibited mRNA expression and protein secretion of transforming growth factor-β (TGF-β), interleukin-33 (IL-33), monocyte chemoattractant protein-1 (MCP-1), and stromal cell-derived factor 1 (SDF-1), which were produced by TNF-α-induced fibroblasts, as measured by quantitative real time-PCR (qRT-PCR) and ELISA assays. The mRNA expression levels of TGF-β, IL-33, SDF-1, and MCP-1 showed 8, 5, 2.5, and 4-fold reductions, respectively. Moreover, secretion of TGF-β, IL-33, SDF-1, and MCP-1 was reduced to 3.65±0.34 ng/mL, 6.3±0.26, 1703.6±295.2, and 5.02±0.18 pg/mL, respectively, compared to the untreated group. In addition, the conditioned media (CM) of TNF-α-stimulated fibroblasts increased the NF-κB expression in colorectal cancer cell lines (HCT-116 and Sw48), while in the vicinity of ZER, the expression of NF-κB was reversed. Considering the significant effects of ZER, this component can be used as an appropriate alternative herbal treatment for cancer-related chronic inflammation.
Collapse
Affiliation(s)
- Zahra Radaei
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 65178, Iran
| | - Alireza Zamani
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 65178, Iran
| | - Rezvan Najafi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, 65178, Iran
| | - Massoud Saidijam
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, 65178, Iran
| | - Farid Azizi Jalilian
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 65178, Iran
| | - Razieh Ezati
- Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, 14965, Iran
| | - Ghasem Solgi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 65178, Iran
| | - Razieh Amini
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, 65178, Iran.
| |
Collapse
|
5
|
Stubbs EB. Targeting the blood-nerve barrier for the management of immune-mediated peripheral neuropathies. Exp Neurol 2020; 331:113385. [PMID: 32562668 DOI: 10.1016/j.expneurol.2020.113385] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/03/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023]
Abstract
Healthy peripheral nerves encounter, with increased frequency, numerous chemical, biological, and biomechanical forces. Over time and with increasing age, these forces collectively contribute to the pathophysiology of a spectrum of traumatic, metabolic, and/or immune-mediated peripheral nerve disorders. The blood-nerve barrier (BNB) serves as a critical first-line defense against chemical and biologic insults while biomechanical forces are continuously buffered by a dense array of longitudinally orientated epineural collagen fibers exhibiting high-tensile strength. As emphasized throughout this Experimental Neurology Special Issue, the BNB is best characterized as a functionally dynamic multicellular vascular unit comprised of not only highly specialized endoneurial endothelial cells, but also associated perineurial cells, pericytes, Schwann cells, basement membrane, and invested axons. The composition of the BNB, while anatomically distinct, is not functionally dissimilar to that of the well characterized neurovascular unit of the central nervous system. While the BNB lacks a glial limitans and an astrocytic endfoot layer, the primary function of both vascular units is to establish, maintain, and protect an optimal endoneurial (PNS) or interstitial (CNS) fluid microenvironment that is vital for proper neuronal function. Altered endoneurial homeostasis as a secondary consequence of BNB dysregulation is considered an early pathological event in the course of a variety of traumatic, immune-mediated, or metabolically acquired peripheral neuropathies. In this review, emerging experimental advancements targeting the endoneurial microvasculature for the therapeutic management of immune-mediated inflammatory peripheral neuropathies, including the AIDP variant of Guillain-Barré syndrome, are discussed.
Collapse
Affiliation(s)
- Evan B Stubbs
- Research Service (151), Department of Veterans Affairs, Edward Hines Jr. VA Hospital, Hines, IL 60141, USA; Department of Ophthalmology, Loyola University Health Science Division, Maywood, IL 60153, USA.
| |
Collapse
|
6
|
Hemshekhar M, Choi KYG, Mookherjee N. Host Defense Peptide LL-37-Mediated Chemoattractant Properties, but Not Anti-Inflammatory Cytokine IL-1RA Production, Is Selectively Controlled by Cdc42 Rho GTPase via G Protein-Coupled Receptors and JNK Mitogen-Activated Protein Kinase. Front Immunol 2018; 9:1871. [PMID: 30158931 PMCID: PMC6104452 DOI: 10.3389/fimmu.2018.01871] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 07/30/2018] [Indexed: 12/20/2022] Open
Abstract
The human host defense peptide LL-37 promotes immune activation such as induction of chemokine production and recruitment of leukocytes. Conversely, LL-37 also mediates anti-inflammatory responses such as production of anti-inflammatory cytokines, e.g., IL-1RA, and the control of pro-inflammatory cytokines, e.g., TNF. The mechanisms regulating these disparate immunomodulatory functions of LL-37 are not completely understood. Rho GTPases are GTP-binding proteins that promote fundamental immune functions such as chemokine production and recruitment of leukocytes. However, recent studies have shown that distinct Rho proteins can both negatively and positively regulate inflammation. Therefore, we interrogated the role of Rho GTPases in LL-37-mediated immunomodulation. We demonstrate that LL-37-induced production of chemokines, e.g., GRO-α and IL-8 is largely dependent on Cdc42/Rac1 Rho GTPase, but independent of the Ras pathway. In contrast, LL-37-induced production of the anti-inflammatory cytokine IL-1RA is not dependent on either Cdc42/Rac1 RhoGTPase or Ras GTPase. Functional studies confirmed that LL-37-induced recruitment of leukocytes (monocytes and neutrophils) is also dependent on Cdc42/Rac1 RhoGTPase activity. We demonstrate that Cdc42/Rac1-dependent bioactivity of LL-37 involves G-protein-coupled receptors (GPCR) and JNK mitogen-activated protein kinase (MAPK) signaling, but not p38 or ERK MAPK signaling. We further show that LL-37 specifically enhances the activity of Cdc42 Rho GTPase, and that the knockdown of Cdc42 suppresses LL-37-induced production of chemokines without altering the peptide's ability to induce IL-1RA. This is the first study to demonstrate the role of Rho GTPases in LL-37-mediated responses. We demonstrate that LL-37 facilitates chemokine production and leukocyte recruitment engaging Cdc42/Rac1 Rho GTPase via GPCR and the JNK MAPK pathway. In contrast, LL-37-mediated anti-inflammatory cytokine IL-1RA production is independent of either Rho or Ras GTPase. The results of this study suggest that Cdc42 Rho GTPase may be the molecular switch that controls the opposing functions of LL-37 in the process of inflammation.
Collapse
Affiliation(s)
- Mahadevappa Hemshekhar
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Ka-Yee Grace Choi
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Neeloffer Mookherjee
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
7
|
Bradbury P, Traini D, Ammit AJ, Young PM, Ong HX. Repurposing of statins via inhalation to treat lung inflammatory conditions. Adv Drug Deliv Rev 2018; 133:93-106. [PMID: 29890243 DOI: 10.1016/j.addr.2018.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/14/2018] [Accepted: 06/06/2018] [Indexed: 12/22/2022]
Abstract
Despite many therapeutic advancements over the past decade, the continued rise in chronic inflammatory lung diseases incidence has driven the need to identify and develop new therapeutic strategies, with superior efficacy to treat these diseases. Statins are one class of drug that could potentially be repurposed as an alternative treatment for chronic lung diseases. They are currently used to treat hypercholesterolemia by inhibiting the 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, that catalyses the rate limiting step in the mevalonate biosynthesis pathway, a key intermediate in cholesterol metabolism. Recent research has identified statins to have other protective pleiotropic properties including anti-inflammatory, anti-oxidant, muco-inhibitory effects that may be beneficial for the treatment of chronic inflammatory lung diseases. However, clinical studies have yielded conflicting results. This review will summarise some of the current evidences for statins pleiotropic effects that could be applied for the treatment of chronic inflammatory lung diseases, their mechanisms of actions, and the potential to repurpose statins as an inhaled therapy, including a detailed discussion on their different physical-chemical properties and how these characteristics could ultimately affect treatment efficacies. The repurposing of statins from conventional anti-cholesterol oral therapy to inhaled anti-inflammatory formulation is promising, as it provides direct delivery to the airways, reduced risk of side effects, increased bioavailability and tailored physical-chemical properties for enhanced efficacy.
Collapse
|
8
|
Li M, Wen Y, Zhang R, Xie F, Zhang G, Qin X. Adenoviral vector-induced silencing of RGMa attenuates blood-brain barrier dysfunction in a rat model of MCAO/reperfusion. Brain Res Bull 2018; 142:54-62. [PMID: 29935233 DOI: 10.1016/j.brainresbull.2018.06.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 06/15/2018] [Accepted: 06/18/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Repulsive guidance molecule A (RGMa) is implicated in focal cerebral ischemia-reperfusion (I/R) injury, but its mechanisms are still largely unknown. This work focused on the effects of RGMa on the blood-brain barrier (BBB) after focal cerebral I/R injury. METHODS Sprague-Dawley (SD) rats were randomly divided into four groups: sham, middle cerebral artery occlusion (MCAO)/reperfusion (I/R), MCAO/reperfusion administered recombinant adenovirus expressing sh-con (I/R + sh-con) and MCAO/reperfusion administered recombinant adenovirus expressing sh-RGMa (I/R + sh-RGMa) groups. Infarct volume, brain edema and neurological scores were evaluated at 3 day after reperfusion. Evens blue leakage and transmission electron microscopy was performed. And the expression level of claudin-5 and ZO-1, CDC-42 and PAK-1, RGMa were detected by western blot. RESULTS Compared with I/R or I/R + sh-con groups, I/R + sh-RGMa group showed smaller infarction volume, attenuated brain edema, improved neurological scores and better BBB integrity, such as reduced Evans blue leakage and ultra-structural change. We also observed improved BBB function followed by down-regulation of MMP-9 and up-regulation of claudin-5 and ZO-1 in the I/R + sh-RGMa group. In addition, up-regulation of the CDC-42 and PAK-1 in the I/R + sh-RGMa group was obtained. CONCLUSIONS RGMa may be involved in I/R injury associated with BBB dysfunction via the CDC-42/PAK-1 signal pathway and may be a promising therapeutic target for I/R injury.
Collapse
Affiliation(s)
- Min Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, Inner Mongolia People's Hospital, Hohhot, China
| | - Yuetao Wen
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Rongrong Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fei Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gang Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
9
|
Bifulco M, Pisanti S, Ciaglia E. The complex interplay among hepatocytes and immune cells at the crossroad between inflammation and cholesterol metabolism in hyperglycemia. Endocrine 2017; 58:21-23. [PMID: 28181143 DOI: 10.1007/s12020-017-1254-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 01/31/2017] [Indexed: 02/05/2023]
Abstract
An intriguing piece of evidence for the inflammatory story of type 2 diabetes has come to the light. A recent study by Okin and colleagues adds new clues to the interplay between cholesterol metabolism and immunity and how it impacts glucose homeostasis in inflammatory conditions. But some questions are a still unsolved conundrum. Here we suggest to better dissect the regulatory mechanism of Mevalonate pathway and to underpin a new causative link between immunometabolic dysregulation and diabetes.
Collapse
Affiliation(s)
- Maurizio Bifulco
- Department of Medicine and Surgery, University of Salerno, Via Salvatore Allende, Baronissi, 84081, Salerno, Italy.
- CORPOREA-Fondazione Idis-Città della Scienza, via Coroglio 104 e 57, 80124, Naples, Italy.
| | - Simona Pisanti
- Department of Medicine and Surgery, University of Salerno, Via Salvatore Allende, Baronissi, 84081, Salerno, Italy
| | - Elena Ciaglia
- Department of Medicine and Surgery, University of Salerno, Via Salvatore Allende, Baronissi, 84081, Salerno, Italy
| |
Collapse
|
10
|
King JR, Gillevet TC, Kabbani N. A G protein-coupled α7 nicotinic receptor regulates signaling and TNF-α release in microglia. FEBS Open Bio 2017; 7:1350-1361. [PMID: 28904864 PMCID: PMC5586346 DOI: 10.1002/2211-5463.12270] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 07/14/2017] [Indexed: 11/21/2022] Open
Abstract
Acetylcholine activation of α7 nicotinic acetylcholine receptors (α7 nAChRs) in microglia attenuates neuroinflammation and regulates TNF‐α release. We used lipopolysaccharide to model inflammation in the microglial cell line EOC20 and examined signaling by the α7 nAChR. Co‐immunoprecipitation experiments confirm that α7 nAChRs bind heterotrimeric G proteins in EOC20 cells. Interaction with Gαi mediates α7 nAChR signaling via enhanced intracellular calcium release and a decrease in cAMP, p38 phosphorylation, and TNF‐α release. These α7 nAChR effects were blocked by the inhibition of Gαi signaling via pertussis toxin, PLC activity with U73122, and α7 nAChR channel activity with the selective antagonist α‐bungarotoxin. Moreover, α7 nAChR signaling in EOC20 cells was significantly diminished by the expression of a dominant‐negative α7 nAChR, α7345‐8A, shown to be impaired in G protein binding. These findings indicate an essential role for G protein coupling in α7 nAChR function in microglia leading to the regulation of inflammation in the nervous system.
Collapse
Affiliation(s)
- Justin R King
- Interdisciplinary Program in Neuroscience Krasnow Institute for Advanced Study George Mason University Fairfax VA USA
| | - Trudy C Gillevet
- Interdisciplinary Program in Neuroscience Krasnow Institute for Advanced Study George Mason University Fairfax VA USA
| | - Nadine Kabbani
- School of Systems Biology Krasnow Institute for Advanced Study George Mason University Fairfax VA USA
| |
Collapse
|
11
|
Zhou X, Wang Y, Wang Y, Cipriano P, Xiao B, Zhou W. Inhibition of triggering receptor expressed on myeloid cells-1 ameliorates experimental autoimmune neuritis. Mol Med Rep 2017; 15:1565-1570. [PMID: 28260057 PMCID: PMC5365016 DOI: 10.3892/mmr.2017.6167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 12/15/2016] [Indexed: 12/30/2022] Open
Abstract
Experimental autoimmune neuritis (EAN) is a cluster of differentiation 4+ T helper 1 cell-mediated inflammatory demyelinating disease of the peripheral nervous system and serves as a useful animal model for Guillain‑Barré syndrome. Triggering receptor expressed on myeloid cells‑1 (TREM‑1) is an important receptor involved in sepsis and the innate inflammatory response. Linear plasmid 17 (LP 17) peptide is a competitive antagonist of TREM‑1. To investigate the role of TREM‑1 in EAN, 64 male Lewis rats were randomly divided into four groups: Normal saline, complete Freund's adjuvant, EAN and LP 17. The present study assessed the mRNA expression levels of TREM‑1, tumor necrosis factor‑α and interleukin‑1β in sciatic nerves and peripheral blood mononuclear cells. The results demonstrated that inhibiting TREM-1 by administering LP 17 ameliorated symptoms and reduced inflammation in EAN rats. The present study concluded that TREM‑1 may be involved in the pathogenesis of EAN, and that inhibition of TREM-1 may ameliorate EAN.
Collapse
Affiliation(s)
- Xiaoliang Zhou
- Department of Neurology, Xiangya Hospital, Central‑South University, Changsha, Hunan 410008, P.R. China
| | - Yanli Wang
- Department of Neurology, Jingzhou Central Hospital, Jingzhou, Hubei 434020, P.R. China
| | - Yuzhong Wang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Peter Cipriano
- Department of Radiology, Stanford Medical Center, Stanford, CA 94305, USA
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central‑South University, Changsha, Hunan 410008, P.R. China
| | - Wenbin Zhou
- Department of Neurology, Xiangya Hospital, Central‑South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
12
|
Langert KA, Goshu B, Stubbs EB. Attenuation of experimental autoimmune neuritis with locally administered lovastatin-encapsulating poly(lactic-co-glycolic) acid nanoparticles. J Neurochem 2016; 140:334-346. [PMID: 27861905 DOI: 10.1111/jnc.13892] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 09/28/2016] [Accepted: 11/01/2016] [Indexed: 12/26/2022]
Abstract
Acute inflammatory demyelinating polyneuropathy (AIDP) is an aggressive antibody- and T-cell-mediated variant of Guillain-Barré Syndrome (GBS), a prominent and debilitating autoimmune disorder of the peripheral nervous system. Despite advancements in clinical management, treatment of patients with AIDP/GBS and its chronic variant CIDP remains palliative and relies on the use of non-specific immunemodulating therapies. Our laboratory has previously reported that therapeutic administration of statins safely attenuates the clinical severity of experimental autoimmune neuritis (EAN), a well-characterized animal model of AIDP/GBS, by restricting the migration of autoreactive leukocytes across peripheral nerve microvascular endoneurial endothelial cells that form the blood-nerve barrier. Despite these advancements, the clinical application of systemically administered statins for the management of inflammatory disorders remains controversial as a result of disappointingly inconclusive phase trials. Here, poly(lactic-co-glycolic) acid (PLGA) nanoparticles were evaluated as an alternative strategy by which to locally administer statins for the management of EAN. When tested in vitro, lovastatin-encapsulating PLGA nanoparticles elicited a marked increase in RhoB mRNA content in peripheral nerve microvascular endoneurial endothelial cells, similar to cells treated with activated unencapsulated lovastatin. Unilateral peri-neural administration of lovastatin-encapsulating PLGA nanoparticles, but not empty nanoparticles, to naïve Lewis rats similarly enhanced RhoB mRNA content in adjacent nerve and muscle tissue. When administered in this manner, serum levels of lovastatin were below the level of detection. Bilateral peri-neural administration of lovastatin-encapsulating PLGA nanoparticles to EAN-induced Lewis rats significantly attenuated EAN clinical severity while protecting against EAN-induced peripheral nerve morphological and functional deficits. This study provides the first proof-of-concept approach for the application of a nanoparticle-based local drug delivery platform for the management of inflammatory demyelinating diseases, including AIDP/GBS.
Collapse
Affiliation(s)
- Kelly A Langert
- Research Service, Department of Veterans Affairs, Edward Hines Jr. VA Hospital, Hines, Illinois, USA
| | - Bruktawit Goshu
- Research Service, Department of Veterans Affairs, Edward Hines Jr. VA Hospital, Hines, Illinois, USA
| | - Evan B Stubbs
- Research Service, Department of Veterans Affairs, Edward Hines Jr. VA Hospital, Hines, Illinois, USA.,Program of Neuroscience, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA.,Department of Ophthalmology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| |
Collapse
|
13
|
Wang Y, Zhang J, Luo P, Zhu J, Feng J, Zhang HL. Tumor necrosis factor-α in Guillain-Barré syndrome, friend or foe? Expert Opin Ther Targets 2016; 21:103-112. [PMID: 27817222 DOI: 10.1080/14728222.2017.1258402] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ying Wang
- Neuroscience Center, Department of Neurology, the First Hospital of Jilin University, Changchun, China
| | - Jingdian Zhang
- Neuroscience Center, Department of Neurology, the First Hospital of Jilin University, Changchun, China
| | - Peijuan Luo
- Norman Bethune Health Science Center, Jilin University, Changchun, China
| | - Jie Zhu
- Neuroscience Center, Department of Neurology, the First Hospital of Jilin University, Changchun, China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Jiachun Feng
- Neuroscience Center, Department of Neurology, the First Hospital of Jilin University, Changchun, China
| | - Hong-Liang Zhang
- Neuroscience Center, Department of Neurology, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Pervan CL, Lautz JD, Blitzer AL, Langert KA, Stubbs EB. Rho GTPase signaling promotes constitutive expression and release of TGF-β2 by human trabecular meshwork cells. Exp Eye Res 2016; 146:95-102. [PMID: 26743044 PMCID: PMC4893883 DOI: 10.1016/j.exer.2015.12.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/15/2015] [Accepted: 12/22/2015] [Indexed: 12/22/2022]
Abstract
Elevated intraocular pressure (IOP) is causally implicated in the pathophysiology of primary open-angle glaucoma (POAG). The molecular mechanisms responsible for elevated IOP remain elusive, but may involve aberrant expression and signaling of transforming growth factor (TGF)-β2 within the trabecular meshwork (TM). Consistent with previously published studies, we show here that exogenous addition of TGF-β2 to cultured porcine anterior segments significantly attenuates outflow facility in a time-dependent manner. By comparison, perfusing segments with a TGFβRI/ALK-5 antagonist (SB-431542) unexpectedly elicited a significant and sustained increase in outflow facility, implicating a role for TM-localized constitutive expression and release of TGF-β2. Consistent with this thesis, cultured primary or transformed (GTM3) quiescent human TM cells were found to constitutively express and secrete measurable amounts of biologically-active TGF-β2. Disrupting monomeric GTPase post-translational prenylation and activation with lovastatin or GGTI-298 markedly reduced constitutive TGF-β2 expression and release. Specifically, inhibiting the Rho subfamily of GTPases with C3 exoenzyme similarly reduced constitutive expression and secretion of TGF-β2. These findings suggest that Rho GTPase signaling, in part, regulates constitutive expression and release of biologically-active TGF-β2 from human TM cells. Localized constitutive expression and release of TGF-β2 by TM cells may promote or exacerbate elevation of IOP in POAG.
Collapse
Affiliation(s)
- Cynthia L Pervan
- Research Service, Department of Veterans Affairs, Edward Hines Jr. VA Hospital, Hines, IL, USA; Department of Ophthalmology, Loyola University Chicago, Maywood, IL, USA.
| | - Jonathan D Lautz
- Research Service, Department of Veterans Affairs, Edward Hines Jr. VA Hospital, Hines, IL, USA; Program in Neuroscience, Loyola University Chicago, Maywood, IL, USA
| | - Andrea L Blitzer
- Research Service, Department of Veterans Affairs, Edward Hines Jr. VA Hospital, Hines, IL, USA; Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Kelly A Langert
- Research Service, Department of Veterans Affairs, Edward Hines Jr. VA Hospital, Hines, IL, USA
| | - Evan B Stubbs
- Research Service, Department of Veterans Affairs, Edward Hines Jr. VA Hospital, Hines, IL, USA; Department of Ophthalmology, Loyola University Chicago, Maywood, IL, USA; Program in Neuroscience, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
15
|
Greathouse KM, Palladino SP, Dong C, Helton ES, Ubogu EE. Modeling leukocyte trafficking at the human blood-nerve barrier in vitro and in vivo geared towards targeted molecular therapies for peripheral neuroinflammation. J Neuroinflammation 2016; 13:3. [PMID: 26732309 PMCID: PMC4702318 DOI: 10.1186/s12974-015-0469-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 12/24/2015] [Indexed: 12/19/2022] Open
Abstract
Peripheral neuroinflammation is characterized by hematogenous mononuclear leukocyte infiltration into peripheral nerves. Despite significant clinical knowledge, advancements in molecular biology and progress in developing specific drugs for inflammatory disorders such as rheumatoid arthritis, inflammatory bowel disease, and multiple sclerosis, there are currently no specific therapies that modulate pathogenic peripheral nerve inflammation. Modeling leukocyte trafficking at the blood-nerve barrier using a reliable human in vitro model and potential intravital microscopy techniques in representative animal models guided by human observational data should facilitate the targeted modulation of the complex inflammatory cascade needed to develop safe and efficacious therapeutics for immune-mediated neuropathies and chronic neuropathic pain.
Collapse
Affiliation(s)
- Kelsey M Greathouse
- Department of Neurology, Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, The University of Alabama at Birmingham, 1825 University Boulevard, Room 1131, Birmingham, AL, 35294-0017, USA.
| | - Steven P Palladino
- Department of Neurology, Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, The University of Alabama at Birmingham, 1825 University Boulevard, Room 1131, Birmingham, AL, 35294-0017, USA.
| | - Chaoling Dong
- Department of Neurology, Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, The University of Alabama at Birmingham, 1825 University Boulevard, Room 1131, Birmingham, AL, 35294-0017, USA.
| | - Eric S Helton
- Department of Neurology, Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, The University of Alabama at Birmingham, 1825 University Boulevard, Room 1131, Birmingham, AL, 35294-0017, USA.
| | - Eroboghene E Ubogu
- Department of Neurology, Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, The University of Alabama at Birmingham, 1825 University Boulevard, Room 1131, Birmingham, AL, 35294-0017, USA.
| |
Collapse
|
16
|
Si MY, Fan ZC, Li YZ, Chang XL, Xie QD, Jiao XY. The prognostic significance of serum and cerebrospinal fluid MMP-9, CCL2 and sVCAM-1 in leukemia CNS metastasis. J Neurooncol 2015; 122:229-44. [PMID: 25630624 DOI: 10.1007/s11060-014-1707-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 12/24/2014] [Indexed: 02/05/2023]
Abstract
Metastasis to the central nervous system (CNS) is the primary obstacle in leukemia treatment. Matrix metalloproteinase-9 (MMP-9), chemokine ligand-2 (CCL2) and soluble vascular adhesion molecule-1 (sVCAM-1) play crucial roles in tumor cell adhesion, motivation and survival, but their roles in leukemia CNS metastasis remain to be elucidated. We investigated the prognostic significance of serum and cerebrospinal fluid (CSF) MMP-9, CCL2 and sVCAM-1 in leukemia patients to explore their potential as predictive biomarkers of the development of CNS leukemia (CNSL). MMP-9, CCL2 and sVCAM-1 were measured in paired CSF and serum samples collecting from 33 leukemia patients with or without CNS metastasis. Other risk factors related to CNSL prognosis were also analyzed. sVCAM-1Serum and CCL2Serum/CSF were significantly higher in the CNSL group than in the non-CNSL group and the controls (p < 0.05). MMP-9Serum was insignificantly lower in the CNSL group than in the non-CNSL group and the controls (p > 0.05). No differences were found for the sVCAM-1Serum, CCL2Serum, and MMP-9Serum levels between non-CNSL patients and controls (p > 0.05). MMP-9CSF was significantly higher in the CNSL group than both the non-CNSL and the control groups (p < 0.05). The indexes of sVCAM-1, CCL2, and MMP-9 in the CNSL group were lower than in the controls (p < 0.05). Positive correlations were determined between the MMP-9CSF and the ALBCSF/BBB value/WBCCSF, between sVCAM-1Serum and the WBCCSF/BBB value. Negative correlations existed between MMP-9Serum and the ALBCSF/BBB value/WBCCSF, and between the CCL2 index and ALBCSF. sVCAM-1Serum was positively associated with event-free survival (EFS), and patients with higher levels of ALBCSF, MMP-9CSF/Serum, CCL2CSF/Serum, and sVCAM-1CSF/Serum had shorter EFS. MMP-9CSF, CCL2CSF and sVCAM-1CSF are the first three principal components analyzed by cluster and principal component analysis. Our data suggest that MMP-9, CCL2 and sVCAM-1 in the CSF may be more potent than serum in predicting the possibility of leukemia metastatic CNS and the outcome of CNSL patients.
Collapse
Affiliation(s)
- Meng-Ya Si
- Department of Cell Biology and Genetics, Shantou University Medical College, 22 Xinling Road, Shantou, 515041, Guangdong, China
| | | | | | | | | | | |
Collapse
|
17
|
Pathogenic mechanisms in inflammatory and paraproteinaemic peripheral neuropathies. Curr Opin Neurol 2014; 27:541-51. [DOI: 10.1097/wco.0000000000000137] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
18
|
Langert KA, Pervan CL, Stubbs EB. Novel role of Cdc42 and RalA GTPases in TNF-α mediated secretion of CCL2. Small GTPases 2014; 5:29260. [PMID: 24911990 DOI: 10.4161/sgtp.29260] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Transendothelial migration of autoreactive leukocytes into peripheral nerves is an early pathological hallmark of acute inflammatory demyelinating polyneuropathy (AIDP), a North American and European variant of Guillain-Barré Syndrome. Whereas the clinical management of AIDP is currently limited to non-selective immune modulating therapies, recent experimental studies support selective targeting of leukocyte trafficking as a promising alternative therapeutic strategy. Here, using a combination of targeted siRNA knockdown and pharmacological inhibitors, we report a novel role of both Cdc42 and RalA GTPases in facilitating TNF-α mediated CCL2 trafficking and release from immortalized rat peripheral nerve microvascular endoneurial endothelial cells. These findings raise interest in Cdc42 and RalA GTPases as potential therapeutic targets for the management of autoimmune inflammatory peripheral nerve disease.
Collapse
Affiliation(s)
- Kelly A Langert
- Research Service; Department of Veterans Affairs; Edward Hines Jr. VA Hospital; Hines, IL USA; Neuroscience Institute; Stritch School of Medicine; Loyola University Chicago; Maywood, IL USA; Department of Ophthalmology; Stritch School of Medicine; Loyola University Chicago; Maywood, IL USA
| | - Cynthia L Pervan
- Research Service; Department of Veterans Affairs; Edward Hines Jr. VA Hospital; Hines, IL USA; Neuroscience Institute; Stritch School of Medicine; Loyola University Chicago; Maywood, IL USA; Department of Ophthalmology; Stritch School of Medicine; Loyola University Chicago; Maywood, IL USA
| | - Evan B Stubbs
- Research Service; Department of Veterans Affairs; Edward Hines Jr. VA Hospital; Hines, IL USA; Neuroscience Institute; Stritch School of Medicine; Loyola University Chicago; Maywood, IL USA; Department of Ophthalmology; Stritch School of Medicine; Loyola University Chicago; Maywood, IL USA
| |
Collapse
|