401
|
Yuan Z, Min J, Zhao Y, Cheng Q, Wang K, Lin S, Luo J, Liu H. Quercetin rescued TNF-alpha-induced impairments in bone marrow-derived mesenchymal stem cell osteogenesis and improved osteoporosis in rats. Am J Transl Res 2018; 10:4313-4321. [PMID: 30662673 PMCID: PMC6325508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 11/24/2018] [Indexed: 06/09/2023]
Abstract
To investigate the effect of quercetin on promoting the proliferation of bone marrow mesenchymal stem cells (BMSCs) and improving osteoporosis in rats. Rats were randomly divided into the sham, OVX and quercetin+OVX groups. In the sham and OVX groups, rats were given carboxymethyl cellulose sodium (CMC-Na). In the quercetin+OVX group, rats were given quercetin (50 mg/kg) once a day. Eight weeks after rats were treated, femurs were subjected to micro-CT scans, and bone biomechanical properties were analysed by the three-point flexural test. In addition, BMSCs were isolated and characterised by MTT, RT-PCR and Western blot analysis. In vivo, quercetin increased bone mineral density (BMD) and improved bone biomechanical properties in postmenopausal osteoporosis rat models. In vitro, TNF-α led to the activation of nuclear factor-kappa B (NF-κB) and the degradation of β-catenin, which were significantly inhibited by quercetin. Furthermore, quercetin promoted BMSC proliferation and osteogenic differentiation. In conclusion, quercetin improved in vitro models of osteoporosis and protected against TNF-α-induced impairments in BMSC osteogenesis.
Collapse
Affiliation(s)
- Zhen Yuan
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang UniversityNanchang, Jiangxi, People’s Republic of China
| | - Jun Min
- Department of Rehabilitation, The Third Affiliated Hospital of Nanchang UniversityNanchang, Jiangxi, People’s Republic of China
| | - Yawen Zhao
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang UniversityNanchang, Jiangxi, People’s Republic of China
| | - Qingfeng Cheng
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang UniversityNanchang, Jiangxi, People’s Republic of China
| | - Kai Wang
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang UniversityNanchang, Jiangxi, People’s Republic of China
| | - Sijian Lin
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang UniversityNanchang, Jiangxi, People’s Republic of China
| | - Jun Luo
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang UniversityNanchang, Jiangxi, People’s Republic of China
| | - Hao Liu
- Department of Rehabilitation, The Second Affiliated Hospital of Nanchang UniversityNanchang, Jiangxi, People’s Republic of China
| |
Collapse
|
402
|
Van Dyken P, Lacoste B. Impact of Metabolic Syndrome on Neuroinflammation and the Blood-Brain Barrier. Front Neurosci 2018; 12:930. [PMID: 30618559 PMCID: PMC6297847 DOI: 10.3389/fnins.2018.00930] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/27/2018] [Indexed: 12/29/2022] Open
Abstract
Metabolic syndrome, which includes diabetes and obesity, is one of the most widespread medical conditions. It induces systemic inflammation, causing far reaching effects on the body that are still being uncovered. Neuropathologies triggered by metabolic syndrome often result from increased permeability of the blood-brain-barrier (BBB). The BBB, a system designed to restrict entry of toxins, immune cells, and pathogens to the brain, is vital for proper neuronal function. Local and systemic inflammation induced by obesity or type 2 diabetes mellitus can cause BBB breakdown, decreased removal of waste, and increased infiltration of immune cells. This leads to disruption of glial and neuronal cells, causing hormonal dysregulation, increased immune sensitivity, or cognitive impairment depending on the affected brain region. Inflammatory effects of metabolic syndrome have been linked to neurodegenerative diseases. In this review, we discuss the effects of obesity and diabetes-induced inflammation on the BBB, the roles played by leptin and insulin resistance, as well as BBB changes occurring at the molecular level. We explore signaling pathways including VEGF, HIFs, PKC, Rho/ROCK, eNOS, and miRNAs. Finally, we discuss the broader implications of neural inflammation, including its connection to Alzheimer's disease, multiple sclerosis, and the gut microbiome.
Collapse
Affiliation(s)
- Peter Van Dyken
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
403
|
Wang Z, Liu CH, Huang S, Chen J. Wnt Signaling in vascular eye diseases. Prog Retin Eye Res 2018; 70:110-133. [PMID: 30513356 DOI: 10.1016/j.preteyeres.2018.11.008] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/21/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022]
Abstract
The Wnt signaling pathway plays a pivotal role in vascular morphogenesis in various organs including the eye. Wnt ligands and receptors are key regulators of ocular angiogenesis both during the eye development and in vascular eye diseases. Wnt signaling participates in regulating multiple vascular beds in the eye including regression of the hyaloid vessels, and development of structured layers of vasculature in the retina. Loss-of-function mutations in Wnt signaling components cause rare genetic eye diseases in humans such as Norrie disease, and familial exudative vitreoretinopathy (FEVR) with defective ocular vasculature. On the other hand, experimental studies in more prevalent vascular eye diseases, such as wet age-related macular degeneration (AMD), diabetic retinopathy (DR), retinopathy of prematurity (ROP), and corneal neovascularization, suggest that aberrantly increased Wnt signaling is one of the causations for pathological ocular neovascularization, indicating the potential of modulating Wnt signaling to ameliorate pathological angiogenesis in eye diseases. This review recapitulates the key roles of the Wnt signaling pathway during ocular vascular development and in vascular eye diseases, and pharmaceutical approaches targeting the Wnt signaling as potential treatment options.
Collapse
Affiliation(s)
- Zhongxiao Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, United States
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, United States
| | - Shuo Huang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, United States
| | - Jing Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, United States.
| |
Collapse
|
404
|
Kuo HY, Liu FC. Molecular Pathology and Pharmacological Treatment of Autism Spectrum Disorder-Like Phenotypes Using Rodent Models. Front Cell Neurosci 2018; 12:422. [PMID: 30524240 PMCID: PMC6262306 DOI: 10.3389/fncel.2018.00422] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/29/2018] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder with a high prevalence rate. The core symptoms of ASD patients are impaired social communication and repetitive behavior. Genetic and environmental factors contribute to pathophysiology of ASD. Regarding environmental risk factors, it is known that valproic acid (VPA) exposure during pregnancy increases the chance of ASD among offspring. Over a decade of animal model studies have shown that maternal treatment with VPA in rodents recapitulates ASD-like pathophysiology at a molecular, cellular and behavioral level. Here, we review the prevailing theories of ASD pathogenesis, including excitatory/inhibitory imbalance, neurotransmitter dysfunction, dysfunction of mTOR and endocannabinoid signaling pathways, neuroinflammation and epigenetic alterations that have been associated with ASD. We also describe the evidence linking neuropathological changes to ASD-like behavioral abnormalities in maternal VPA-treated rodents. In addition to obtaining an understanding of the neuropathological mechanisms, the VPA-induced ASD-like animal models also serve as a good platform for testing pharmacological reagents that might be use treating ASD. We therefore have summarized the various pharmacological studies that have targeted the classical neurotransmitter systems, the endocannabinoids, the Wnt signal pathway and neuroinflammation. These approaches have been shown to often be able to ameliorate the ASD-like phenotypes induced by maternal VPA treatments.
Collapse
Affiliation(s)
- Hsiao-Ying Kuo
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
| | - Fu-Chin Liu
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
405
|
Au DT, Migliorini M, Strickland DK, Muratoglu SC. Macrophage LRP1 Promotes Diet-Induced Hepatic Inflammation and Metabolic Dysfunction by Modulating Wnt Signaling. Mediators Inflamm 2018; 2018:7902841. [PMID: 30524198 PMCID: PMC6247401 DOI: 10.1155/2018/7902841] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 08/22/2018] [Accepted: 09/03/2018] [Indexed: 12/20/2022] Open
Abstract
Hepatic inflammation is associated with the development of insulin resistance, which can perpetuate the disease state and may increase the risk of metabolic syndrome and diabetes. Despite recent advances, mechanisms linking hepatic inflammation and insulin resistance are still unclear. The low-density lipoprotein receptor-related protein 1 (LRP1) is a large endocytic and signaling receptor that is highly expressed in macrophages, adipocytes, hepatocytes, and vascular smooth muscle cells. To investigate the potential role of macrophage LRP1 in hepatic inflammation and insulin resistance, we conducted experiments using macrophage-specific LRP1-deficient mice (macLRP1-/- ) generated on a low-density lipoprotein receptor knockout (LDLR-/- ) background and fed a Western diet. LDLR-/-; macLRP1-/- mice gained less body weight and had improved glucose tolerance compared to LDLR-/- mice. Livers from LDLR-/-; macLRP1-/- mice displayed lower levels of gene expression for several inflammatory cytokines, including Ccl3, Ccl4, Ccl8, Ccr1, Ccr2, Cxcl9, and Tnf, and reduced phosphorylation of GSK3α and p38 MAPK proteins. Furthermore, LRP1-deficient peritoneal macrophages displayed altered cholesterol metabolism. Finally, circulating levels of sFRP-5, a potent anti-inflammatory adipokine that functions as a decoy receptor for Wnt5a, were elevated in LDLR-/-; macLRP1-/- mice. Surface plasmon resonance experiments revealed that sFRP-5 is a novel high affinity ligand for LRP1, revealing that LRP1 regulates levels of this inhibitor of Wnt5a-mediated signaling. Collectively, our results suggest that LRP1 expression in macrophages promotes hepatic inflammation and the development of glucose intolerance and insulin resistance by modulating Wnt signaling.
Collapse
Affiliation(s)
- Dianaly T. Au
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mary Migliorini
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dudley K. Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Selen C. Muratoglu
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
406
|
Hombrebueno JR, Ali IHA, Ma JX, Chen M, Xu H. Antagonising Wnt/β-catenin signalling ameliorates lens-capsulotomy-induced retinal degeneration in a mouse model of diabetes. Diabetologia 2018; 61:2433-2446. [PMID: 30019207 PMCID: PMC6182657 DOI: 10.1007/s00125-018-4682-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/22/2018] [Indexed: 12/15/2022]
Abstract
AIMS/HYPOTHESIS Cataract surgery in diabetic individuals worsens pre-existing retinopathy and triggers the development of diabetic ocular complications, although the underlying cellular and molecular pathophysiology remains elusive. We hypothesise that lens surgery may exaggerate pre-existing retinal inflammation in diabetes, which may accelerate neurovascular degeneration in diabetic eyes. METHODS Male heterozygous Ins2Akita mice (3 months of age) and C57BL/6 J age-matched siblings received either lens capsulotomy (to mimic human cataract surgery) or corneal incision (sham surgery) in the right eye. At different days post surgery, inflammation in anterior/posterior ocular tissues was assessed by immunohistochemistry and proinflammatory gene expression in the retina by quantitative PCR (qPCR). Degenerative changes in the retina were evaluated by electroretinography, in vivo examination of retinal thickness (using spectral domain optical coherence tomography [SD-OCT]) and morphometric analysis of retinal neurons. The therapeutic benefit of neutralising Wnt/β-catenin signalling following lens capsulotomy was evaluated by intravitreal administration of monoclonal antibody against the co-receptor low-density lipoprotein receptor-related protein 6 (LRP6) (Mab2F1; 5 μg/μl in each eye). RESULTS Lens capsulotomy triggered the early onset of retinal neurodegeneration in Ins2Akita mice, evidenced by abnormal scotopic a- and b-wave responses, reduced retinal thickness and degeneration of outer/inner retinal neurons. Diabetic Ins2Akita mice also had a higher number of infiltrating ionised calcium-binding adapter molecule 1 (IBA1)/CD68+ cells in the anterior/posterior ocular tissues and increased retinal expression of inflammatory mediators (chemokine [C-C motif] ligand 2 [CCL2] and IL-1β). The expression of β-catenin was significantly increased in the inner nuclear layer, ganglion cells and infiltrating immune cells in Ins2Akita mice receiving capsulotomy. Neutralisation of Wnt/β-catenin signalling by Mab2F1 ameliorated ocular inflammation and prevented capsulotomy-induced retinal degeneration in the Ins2Akita mouse model of diabetes. CONCLUSIONS/INTERPRETATION Targeting the canonical Wnt/β-catenin signalling pathway may provide a novel approach for the postoperative management of diabetic individuals needing cataract surgery.
Collapse
Affiliation(s)
- Jose R Hombrebueno
- Centre for Experimental Medicine, Wellcome-Wolfson Institute of Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| | - Imran H A Ali
- Centre for Experimental Medicine, Wellcome-Wolfson Institute of Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Jian-Xing Ma
- Department of Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Mei Chen
- Centre for Experimental Medicine, Wellcome-Wolfson Institute of Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| | - Heping Xu
- Centre for Experimental Medicine, Wellcome-Wolfson Institute of Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| |
Collapse
|
407
|
Cai Z, Feng Y, Li C, Yang K, Sun T, Xu L, Chen Y, Yan CH, Lu WW, Chiu KY. Magnoflorine with hyaluronic acid gel promotes subchondral bone regeneration and attenuates cartilage degeneration in early osteoarthritis. Bone 2018; 116:266-278. [PMID: 30149068 DOI: 10.1016/j.bone.2018.08.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/15/2018] [Accepted: 08/22/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate efficacy of Chinese medicine magnoflorine combined with hyaluronic acid (HA)-gel in promoting subchondral bone (SCB) regeneration and attenuating cartilage degeneration in early osteoarthritis (OA). METHODS MC3T3-E1 under magnoflorine treatment was assayed by XTT to determine cell viability. Cell proliferation was reflected through cell cycle. Osteoblast mineralization was stained by Alizarin Red. Standardized bone canal of 1 mm in diameter and 4 mm in depth was made on tibial medial plateau of 4-month-old Dunkin-Hartley spontaneous knee OA guinea pigs. Guinea pigs (n = 5/group) were treated once intra-bone canal injection of 2 μl HA-gel, 2 μl HA-gel+50 ng magnoflorine and null (Defect) respectively, except sham group. The left hind limbs were harvested for μCT scan and histopathological staining 2-month post-surgery. RESULTS 25 μg/ml magnoflorine treatment significantly increased cell viability, S-phase and mineralization of MC3T3-E1 cells. In vivo, HA-gel + magnoflorine treatment significantly altered SCB microstructure; changes included increase in bone volume fraction (BV/TV), trabecular number (Tb.N), connectivity density (Conn.Dn), and decrease in degree of anisotropy (DA), which implied trabecular bone regeneration. Treatment also resulted in a decrease in modified Mankin's scores, and an increase in volume ratio of hyaline cartilage (HC)/calcified cartilage (CC) and fractal dimension (FD, roughness indicator of osteochondral conjunction), compared to Defect and HA groups. Furthermore, FD was positively associated with volume ratio of HC/CC and negatively associated with modified Mankin's scores. Finally, histological results showed that due to a faster regeneration of SCB with the HA-gel + magnoflorine treatment, the reduction of cartilage matrix and the decreased expression of chondrogenic signals were attenuated. CONCLUSION Our study elucidated the potential benefits of HA-gel + magnoflorine in promoting SCB regeneration and revealed a protective effect of stimulating recovery of the SCB integrity on attenuating cartilage degradation to prevent OA progression.
Collapse
Affiliation(s)
- Zhe Cai
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Yu Feng
- Department of Traumatology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Chentian Li
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Kedi Yang
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Tianhao Sun
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Lei Xu
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Yan Chen
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Chun-Hoi Yan
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - William Weijia Lu
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, China.
| | - Kwong-Yuen Chiu
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
| |
Collapse
|
408
|
Abnormal activation of the Akt signaling pathway in adenoid cystic carcinoma. Eur Arch Otorhinolaryngol 2018; 275:3039-3047. [PMID: 30367261 DOI: 10.1007/s00405-018-5182-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 10/22/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE Adenoid cystic carcinoma (ACC) is an intriguing lesion because it shows a slow growth in the beginning, but a late poor prognosis due to perineural invasion, metastasis and recurrence. This study aimed to investigate whether Akt signaling would be deregulated in adenoid cystic carcinoma and its consequence in the expression of associated proteins. METHODS The expression of the Akt, p-Akt, NFκB, β-catenin, cyclin D1 and COX-2 was assessed by immunohistochemistry in 10 cases of ACC, 17 cases of pleomorphic adenoma (PA), and 7 cases of normal salivary gland (NSG). RESULTS p-Akt was overexpressed in ACC when compared to NSG. NFκB, β-catenin, and COX-2 were overexpressed in ACC and PA when compared to NSG. Most proteins were slightly higher expressed in ACC than in PA, but they never reached significance. p-Akt expression positively correlated with NFκB, β-catenin, cyclin D1 and COX-2 in ACC and PA, while this correlation trended to be negative in for these proteins (except for NFκB) in NSG using Person's correlation analysis, but without reaching significance. CONCLUSIONS Our results indicate an abnormal activation of Akt signaling pathway, which can be an important regulator of tumor biology in ACC. Activated Akt correlated with the expression of NFκB, β-catenin and COX-2, which can potentially influence cell survival in ACC.
Collapse
|
409
|
Abdi J, Rashedi I, Keating A. Concise Review: TLR Pathway-miRNA Interplay in Mesenchymal Stromal Cells: Regulatory Roles and Therapeutic Directions. Stem Cells 2018; 36:1655-1662. [PMID: 30171669 DOI: 10.1002/stem.2902] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/13/2018] [Accepted: 08/08/2018] [Indexed: 12/12/2022]
Abstract
Mesenchymal stromal cells (MSCs) deploy Toll-like receptors (TLRs) to respond to exogenous and endogenous signals. Activation of TLR pathways in MSCs alters their inflammatory profile and immunomodulatory effects on cells from both the innate and adaptive immune systems. Micro-RNAs (miRNAs), whose expression is modulated by TLR activation, can regulate inflammatory responses by targeting components of the TLR signaling pathways either in MSCs or in the cells with which they interact. Here, we review how the miRNA-TLR pathway axis can regulate the immunomodulatory functions of MSCs, including their interactions with monocytes/macrophages and natural killer cells, and discuss the therapeutic implications for MSC-based therapies. Stem Cells 2018;36:1655-1662.
Collapse
Affiliation(s)
- Jahangir Abdi
- Cell Therapy Translational Research Laboratory, University Health Network (UHN), Toronto, Ontario, Canada.,Arthritis Program, Krembil Research Institute, UHN, Toronto, ON, Canada
| | - Iran Rashedi
- Cell Therapy Translational Research Laboratory, University Health Network (UHN), Toronto, Ontario, Canada.,Arthritis Program, Krembil Research Institute, UHN, Toronto, ON, Canada
| | - Armand Keating
- Cell Therapy Translational Research Laboratory, University Health Network (UHN), Toronto, Ontario, Canada.,Arthritis Program, Krembil Research Institute, UHN, Toronto, ON, Canada.,Princess Margaret Cancer Centre, UHN, Toronto, ON, Canada
| |
Collapse
|
410
|
Chen C, Bao GF, Xu G, Sun Y, Cui ZM. Altered Wnt and NF-κB Signaling in Facet Joint Osteoarthritis: Insights from RNA Deep Sequencing. TOHOKU J EXP MED 2018; 245:69-77. [PMID: 29806631 DOI: 10.1620/tjem.245.69] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Facet joint osteoarthritis is common lumbar osteoarthritis characterized by facet joint cartilage degeneration. However, the molecular basis of facet joint osteoarthritis remains largely undetermined. In the current study, we collected facet joint tissue samples from 10 control patients and 48 patients with facet joint osteoarthritis (20 patients with moderate degeneration and 28 with severe degeneration). The control patients underwent internal fixation of the lumbar spine due to vertebral fracture. RNA deep sequencing was performed, and Bioinformatic tools were applied. Among top 30 enriched signaling pathways, we focused on two inflammation-related signaling pathways, Wnt and NF-κB signaling pathways. Subsequently, using the quantitative RT-PCR analysis, we confirmed that in Wnt signaling pathway, the mRNA levels of Dickkopf WNT Signaling Pathway Inhibitor 2 (DKK2), Sex-determining Region Y-box 17 (SOX17), MYC, Cyclin D1, Calcium/Calmodulin Dependent Protein Kinase II Alpha (CAMK2A), and Wnt Family Member 11 and 5 were increased in facet joint osteoarthritis, while the mRNA levels of WNT Inhibitory Factor 1, Casein Kinase 1 Alpha 1, Transcription Factor 7/Lymphoid Enhancer Binding Factor 1 (TCF7/LEF1), and VANGL Planar Cell Polarity Protein 2 were decreased. In NF-κB signaling pathway, the mRNA levels of C-C Motif Chemokine Ligand 4 (CCL4) and C-C Motif Chemokine Ligand 4 Like 2 (CCL4L2) were increased, while the mRNA levels of BCL2 Related Protein A1 were decreased. These results suggest that Wnt and NF-κB signaling may be altered in the process of facet joint cartilage degeneration. The present study will expand our understanding of the molecular bases underlying facet joint osteoarthritis.
Collapse
Affiliation(s)
- Chu Chen
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University
| | - Guo-Feng Bao
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University
| | - Guanhua Xu
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University
| | - Yuyu Sun
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University
| | - Zhi-Ming Cui
- Department of Spine Surgery, The Second Affiliated Hospital of Nantong University
| |
Collapse
|
411
|
Ferranti EM, Aloqaily BH, Gifford CA, Löest CA, Wenzel JC, Hernandez Gifford JA. Lipopolysaccharide modulation of ovarian hormonal profile1. Transl Anim Sci 2018; 2:S31-S34. [PMID: 32704732 PMCID: PMC7200988 DOI: 10.1093/tas/txy027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 04/14/2018] [Indexed: 12/01/2022] Open
Affiliation(s)
- Emily M Ferranti
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM
| | - Bahaa H Aloqaily
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM
| | - Craig A Gifford
- Department of Extension Animal Sciences and Natural Resources, Extension Animal Sciences and Natural Resources, New Mexico State University, Las Cruces, NM
| | - Clint A Löest
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM
| | - John C Wenzel
- Department of Extension Animal Sciences and Natural Resources, Extension Animal Sciences and Natural Resources, New Mexico State University, Las Cruces, NM
| | | |
Collapse
|
412
|
Luo CW, Hsiao IL, Wang JY, Wu CC, Hung WC, Lin YH, Chen TY, Hsu YC, Cheng TL, Pan MR. Cell Motility Facilitated by Mono(2-ethylhexyl) Phthalate via Activation of the AKT-β-Catenin-IL-8 Axis in Colorectal Cancer. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:9635-9644. [PMID: 30188700 DOI: 10.1021/acs.jafc.8b03558] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a common plasticizer that is widely used in many consumer products and medical devices. Humans can be exposed to DEHP through ingestion, inhalation, or dermal absorption. Previous studies on DEHP have focused on its role as an endocrine-disrupting chemical leading to endocrine-related diseases. However, the correlation between DEHP exposure and the progression of colorectal cancer (CRC) is largely unknown. The aim of this study was to investigate the effects of mono(2-ethylhexyl) phthalate (MEHP), an active metabolite of DEHP, on the progression of CRC. Our results showed that treatment with MEHP enriched the population of cancer-stem-cell (CSC)-like cells and upregulated IL-8 expression by inducing the AKT-β-catenin-TCF4 signaling pathway. Blocking β-catenin-TCF4-mediated IL-8 expression reversed the MEHP-induced migration and enrichment of CSC-like cells. Consistent with the in vitro data, DEHP treatment increased the levels of nuclear β-catenin, polyp formation, and invasive adenocarcinoma in a mouse model. Our results suggest that MEHP facilitates the progression of CRC through AKT-β-catenin signaling.
Collapse
Affiliation(s)
- Chi-Wen Luo
- Division of Cardiology , Chang Gung Memorial Hospital, Kaohsiung Medical Center , Kaohsiung 833 , Taiwan
| | - I-Ling Hsiao
- Graduate Institute of Clinical Medicine , Kaohsiung Medical University , Number 100, Tzyou First Road , Kaohsiung 807 , Taiwan
| | - Jaw-Yuan Wang
- Graduate Institute of Clinical Medicine , Kaohsiung Medical University , Number 100, Tzyou First Road , Kaohsiung 807 , Taiwan
- Division of Colorectal Surgery, Department of Surgery , Kaohsiung Medical University Hospital, Kaohsiung Medical University , Kaohsiung 807 , Taiwan
| | - Chun-Chieh Wu
- Department of Pathology , Kaohsiung Medical University Hospital, Kaohsiung Medical University , Kaohsiung 807 , Taiwan
| | - Wen-Chun Hung
- National Institute of Cancer Research , National Health Research Institutes , Tainan 704 , Taiwan
| | - Yu-Han Lin
- Graduate Institute of Clinical Medicine , Kaohsiung Medical University , Number 100, Tzyou First Road , Kaohsiung 807 , Taiwan
| | - Tzu-Yi Chen
- Graduate Institute of Clinical Medicine , Kaohsiung Medical University , Number 100, Tzyou First Road , Kaohsiung 807 , Taiwan
| | - Yin-Chou Hsu
- Graduate Institute of Clinical Medicine , Kaohsiung Medical University , Number 100, Tzyou First Road , Kaohsiung 807 , Taiwan
- Department of Emergency Medicine , E-Da Hospital, I-Shou University , Kaohsiung 824 , Taiwan
| | - Tian-Lu Cheng
- Center for Biomarkers and Biotech Drugs , Kaohsiung Medical University , Kaohsiung 807 , Taiwan
- Department of Biomedical Science and Environmental Biology , Kaohsiung Medical University , Kaohsiung 807 , Taiwan
- Institute of Biomedical Sciences , National Sun Yat-sen University , Kaohsiung 804 , Taiwan
| | - Mei-Ren Pan
- Graduate Institute of Clinical Medicine , Kaohsiung Medical University , Number 100, Tzyou First Road , Kaohsiung 807 , Taiwan
| |
Collapse
|
413
|
Mills KJ, Robinson MK, Sherrill JD, Schnell DJ, Xu J. Analysis of gene expression profiles of multiple skin diseases identifies a conserved signature of disrupted homeostasis. Exp Dermatol 2018; 27:1000-1008. [DOI: 10.1111/exd.13694] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Kevin J. Mills
- The Procter and Gamble Company; Mason Business Center; Mason OH USA
| | | | | | - Daniel J. Schnell
- Department of Biomedical Informatics; Cincinnati Children's Hospital Medical Center; Cincinnati OH USA
| | - Jun Xu
- Calico, LLC; South San Francisco CA USA
| |
Collapse
|
414
|
Ma C, Zhou X, Xu K, Wang L, Yang Y, Wang W, Liu A, Ran J, Yan S, Wu H, Wu L. Specnuezhenide Decreases Interleukin-1β-Induced Inflammation in Rat Chondrocytes and Reduces Joint Destruction in Osteoarthritic Rats. Front Pharmacol 2018; 9:700. [PMID: 30050432 PMCID: PMC6052343 DOI: 10.3389/fphar.2018.00700] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/08/2018] [Indexed: 11/16/2022] Open
Abstract
As a chronic disease, osteoarthritis (OA) leads to the degradation of both cartilage and subchondral bone, its development being mediated by proinflammatory cytokines like interleukin-1β. In the present study, the anti-inflammatory effect of specnuezhenide (SPN) in OA and its underlying mechanism were studied in vitro and in vivo. The results showed that SPN decreases the expression of cartilage matrix-degrading enzymes and the activation of NF-κB and wnt/β-catenin signaling, and increases chondrocyte-specific gene expression in IL-1β-induced inflammation in chondrocytes. Furthermore, SPN treatment prevents the degeneration of both cartilage and subchondral bone in a rat model of OA. To the best of our knowledge, this study is the first to report that SPN decreases interleukin-1β-induced inflammation in rat chondrocytes by inhibiting the activation of the NF-κB and wnt/β-catenin pathways, and, thus, has therapeutic potential in the treatment of OA.
Collapse
Affiliation(s)
- Chiyuan Ma
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaopeng Zhou
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Xu
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Linyan Wang
- Department of Ophthalmology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yute Yang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Wang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - An Liu
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jisheng Ran
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shigui Yan
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haobo Wu
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lidong Wu
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
415
|
Xiong Y, Li KX, Wei H, Jiao L, Yu SY, Zeng L. Eph/ephrin signalling serves a bidirectional role in lipopolysaccharide‑induced intestinal injury. Mol Med Rep 2018; 18:2171-2181. [PMID: 29901151 PMCID: PMC6072232 DOI: 10.3892/mmr.2018.9169] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
A growing body of evidence has demonstrated that Eph/ephrin signalling may serve a central role in intestinal diseases. However, whether erythropoietin-producing hepatocellular (Eph)/ephrin signalling is associated with the development of post-infectious irritable bowel syndrome (PI-IBS) is still unknown. In the present study, the role of Eph/Ephrin signalling in lipopolysaccharide (LPS)-induced intestinal injury was evaluated in vivo and in vitro. LPS treatment significantly increased the levels of proinflammatory mediators [monocyte chemoattractant protein-1, tumour necrosis factor α, interleukin (IL)-1β, IL-6, intercellular adhesion molecule 1 and vascular cell adhesion molecule-1], activated the EphA2-Ephrin A1, protein kinase B (Akt)-nuclear factor (NF)-κB, Src-NF-κB and Wnt/β-catenin signalling pathways, and inhibited EphB1-Ephrin B3 signalling in colon tissues, and primary cultured enteric neuronal and glial cells. Notably, EphA2 monoclonal antibody (mAb) treatment or Ephrin B3 overexpression could partially alleviate the LPS-induced upregulation of proinflammatory mediators, and Akt-NF-κB, Src-NF-κB and Wnt/β-catenin signalling pathways. In addition, EphA2 mAb treatment could partially inhibit LPS-induced inactivation of EphB-Ephrin B3 signalling, while Ephrin B3 overexpression could abrogate LPS-induced activation of EphA2-Ephrin A1 signalling. EphB1/Ephrin B3 signalling may antagonise the EphA2/Ephrin A1-dependent pathway following LPS treatment. The results associated with the EphA2 signaling pathway, indicated that Eph/ephrin signalling may serve a bidirectional role in LPS-induced intestinal injury. Eph/ephrin signalling may be a novel therapeutic target for LPS-induced intestinal injury and potentially PI-IBS.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Gastroenterology, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong 518110, P.R. China
| | - Kai-Xue Li
- Department of Gastroenterology, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| | - Hong Wei
- Department of Gastroenterology, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| | - Lu Jiao
- Department of Gastroenterology, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| | - Shao-Yong Yu
- Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, MD 21205‑2195, USA
| | - Li Zeng
- Department of Gastroenterology, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518035, P.R. China
| |
Collapse
|
416
|
Liu X, Zhang Z, Pan S, Shang S, Li C. Interaction between the Wnt/β-catenin signaling pathway and the EMMPRIN/MMP-2, 9 route in periodontitis. J Periodontal Res 2018; 53:842-852. [PMID: 29900539 DOI: 10.1111/jre.12574] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2018] [Indexed: 12/13/2022]
Affiliation(s)
- X. Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST); Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan Hubei China
| | - Z. Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST); Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan Hubei China
| | - S. Pan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST); Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan Hubei China
| | - S. Shang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST); Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan Hubei China
- Department of Periodontology; School and Hospital of Stomatology; Wuhan University; Wuhan Hubei China
| | - C. Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST); Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan Hubei China
- Department of Periodontology; School and Hospital of Stomatology; Wuhan University; Wuhan Hubei China
| |
Collapse
|
417
|
Chi YT, Xu CJ, Sun XJ, Li DM, Wang HF, Wang MM, He XF. [Expression and significance of secreted frizzled-related protein 1 and β-catenin in gingival tissue of patients with chronic periodontitis]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2018; 36:257-261. [PMID: 29984924 PMCID: PMC7030293 DOI: 10.7518/hxkq.2018.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/01/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE This study aimed to investigate the expression and correlation of secreted frizzled-related protein 1 (SFRP1) and β-catenin in gingival tissues of patients with chronic periodontitis (CP). The role of the classical Wnt/β-catenin signaling pathway in the development of periodontitis was also explored. METHODS Twenty-eight patients with CP (CP group) were enrolled in this study. Among them, 16 cases were moderate CP, and 12 demonstrated severe CP. Twelve healthy cases comprised the controls (normal group). Gingival tissue was collected, and the probing depth, bleeding index, and clinical attachment loss were recorded. The expression levels of SFRP1 and β-catenin were detected by immunohistochemistry, and staining intensity was evaluated by double scoring method. SPSS 19.0 was used for statistical analysis. RESULTS The staining strength scores of SFRP1 and β-catenin were 2.16±0.65 and 1.12±0.51 in the normal group, 3.57±0.45 and 2.36±0.49 in the CP group, 3.61±0.40 and 2.30±0.44 in the moderate CP group, and 3.52±0.52 and 2.45±0.55 in the severe CP group, respectively. The expression of SFRP1 and β-catenin in the CP group was higher than that in the normal group (P<0.01). A significant difference was noted between the normal group and the moderate and severe CP groups (P<0.01) but none between the moderate and severe CP groups (P>0.05). A positive correlation was found between the expression of SFRP1 and β-catenin (r=0.657, P<0.01). The expression levels of β-catenin and SFRP1 were related to periodontal indexes. The correlation between the expression of SFRP1 and probing depth was most significant (r=0.723, P<0.01), as well as that between β-catenin and bleeding index (r=0.697, P<0.01). CONCLUSIONS Patients with CP exhibit elevated expression of SFRP1 and β-catenin in gingival tissues, and this event is related to the degree of periodontal destruction. Abnormal expression of SFRP1 and β-catenin may promote the development of periodontitis.
Collapse
Affiliation(s)
- Yu-Tan Chi
- Center of Stomatology, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Chun-Jiao Xu
- Center of Stomatology, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Xiao-Juan Sun
- Center of Stomatology, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Dong-Mei Li
- Center of Stomatology, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Hong-Feng Wang
- Center of Stomatology, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Miao-Miao Wang
- Center of Stomatology, Xiangya Hospital of Central South University, Changsha 410008, China
| | - Xiu-Fang He
- Center of Stomatology, Xiangya Hospital of Central South University, Changsha 410008, China
| |
Collapse
|
418
|
Natural scaffolds in anticancer therapy and precision medicine. Biotechnol Adv 2018; 36:1563-1585. [PMID: 29729870 DOI: 10.1016/j.biotechadv.2018.04.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 04/08/2018] [Accepted: 04/26/2018] [Indexed: 12/21/2022]
Abstract
The diversity of natural compounds is essential for their mechanism of action. The source, structures and structure activity relationship of natural compounds contributed to the development of new classes of chemotherapy agents for over 40 years. The availability of combinatorial chemistry and high-throughput screening has fueled the challenge to identify novel compounds that mimic nature's chemistry and to predict their macromolecular targets. Combining conventional and targeted therapies helped to successfully overcome drug resistance and prolong disease-free survival. Here, we aim to provide an overview of preclinical investigated natural compounds alone and in combination to further improve personalization of cancer treatment.
Collapse
|
419
|
Vallée A, Vallée JN, Guillevin R, Lecarpentier Y. Interactions Between the Canonical WNT/Beta-Catenin Pathway and PPAR Gamma on Neuroinflammation, Demyelination, and Remyelination in Multiple Sclerosis. Cell Mol Neurobiol 2018; 38:783-795. [PMID: 28905149 PMCID: PMC11482031 DOI: 10.1007/s10571-017-0550-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/09/2017] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is marked by neuroinflammation and demyelination with loss of oligodendrocytes in the central nervous system. The immune response is regulated by WNT/beta-catenin pathway in MS. Activated NF-kappaB, a major effector of neuroinflammation, and upregulated canonical WNT/beta-catenin pathway positively regulate each other. Demyelinating events present an upregulation of WNT/beta-catenin pathway, whereas proper myelinating phases show a downregulation of WNT/beta-catenin pathway essential for the promotion of oligodendrocytes precursors cells proliferation and differentiation. The activation of WNT/beta-catenin pathway results in differentiation failure and impairment in remyelination. However, PI3K/Akt pathway and TCF7L2, two downstream targets of WNT/beta-catenin pathway, are upregulated and promote proper remyelination. The interactions of these signaling pathways remain unclear. PPAR gamma activation can inhibit NF-kappaB, and can also downregulate the WNT/beta-catenin pathway. PPAR gamma and canonical WNT/beta-catenin pathway act in an opposite manner. PPAR gamma agonists appear as a promising treatment for the inhibition of demyelination and the promotion of proper remyelination through the control of both NF-kappaB activity and canonical WNT/beta-catenin pathway.
Collapse
Affiliation(s)
- Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, Poitiers, France.
- Laboratory of Mathematics and Applications (LMA), UMR CNRS 7348, University of Poitiers, Poitiers, France.
| | - Jean-Noël Vallée
- Laboratory of Mathematics and Applications (LMA), UMR CNRS 7348, University of Poitiers, Poitiers, France
- CHU Amiens Picardie, University of Picardie Jules Verne (UPJV), Amiens, France
| | - Rémy Guillevin
- DACTIM, UMR CNRS 7348, University of Poitiers et CHU de Poitiers, Poitiers, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| |
Collapse
|
420
|
L'Episcopo F, Tirolo C, Peruzzotti-Jametti L, Serapide MF, Testa N, Caniglia S, Balzarotti B, Pluchino S, Marchetti B. Neural Stem Cell Grafts Promote Astroglia-Driven Neurorestoration in the Aged Parkinsonian Brain via Wnt/β-Catenin Signaling. Stem Cells 2018; 36:1179-1197. [PMID: 29575325 DOI: 10.1002/stem.2827] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 02/19/2018] [Accepted: 03/06/2018] [Indexed: 12/16/2022]
Abstract
neuronal phenotype. Wnt/β-catenin signaling antagonism abolished mDA neurorestoration and immune modulatory effects of NSC grafts. Our work implicates an unprecedented therapeutic potential for somatic NSC grafts in the restoration of mDA neuronal function in the aged Parkinsonian brain. Stem Cells 2018;36:1179-1197.
Collapse
Affiliation(s)
| | | | - Luca Peruzzotti-Jametti
- Dept of Clinical Neurosciences, Clifford Allbutt Building - Cambridge Biosciences Campus and NIHR Biomedical Research,Centre, University of Cambridge, Hills Road, CB2 0HA Cambridge, UK
| | - Maria F Serapide
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Pharmacology and Physiology Sections, University of Catania Medical School, Catania, Italy
| | | | | | - Beatrice Balzarotti
- Dept of Clinical Neurosciences, Clifford Allbutt Building - Cambridge Biosciences Campus and NIHR Biomedical Research,Centre, University of Cambridge, Hills Road, CB2 0HA Cambridge, UK
| | - Stefano Pluchino
- Dept of Clinical Neurosciences, Clifford Allbutt Building - Cambridge Biosciences Campus and NIHR Biomedical Research,Centre, University of Cambridge, Hills Road, CB2 0HA Cambridge, UK
| | - Bianca Marchetti
- Oasi Research Institute-IRCCS, Troina, Italy.,Department of Biomedical and Biotechnological Sciences (BIOMETEC), Pharmacology and Physiology Sections, University of Catania Medical School, Catania, Italy
| |
Collapse
|
421
|
Wang H, Yao L, Gong Y, Zhang B. TRIM31 regulates chronic inflammation via NF-κB signal pathway to promote invasion and metastasis in colorectal cancer. Am J Transl Res 2018; 10:1247-1259. [PMID: 29736218 PMCID: PMC5934584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 03/07/2018] [Indexed: 06/08/2023]
Abstract
Tripartite motif 31(TRIM31) is a new member of the E3 ubiquitin ligase family, which plays a role in many biological processes. It has been indicated that TRIM31 is strongly correlated with tumorigenesis. However, the impact of TRIM31 in colorectal cancer (CRC) and its underlying mechanisms are largely unknown. In this study, we found an increase in TRIM31 expression in CRC cells and a positive correlation between TRIM31 and CRC malignancy. Applying genetic interventions and cellular function tests, our results showed that TRIM31 acted as an epithelial-mesenchymal transition (EMT) inducer to promote CRC invasion and metastasis. Mechanical exploration revealed that TRIM31 mediated the upregulation of inflammatory cytokines TNF, IL-1β, and IL-6 through the NF-κB pathway, thus contributing to EMT in CRC progression. Collectively, we concluded that TRIM31 mediated chronic inflammation via the NF-κB pathway to promote invasion and metastasis in CRC. Therefore, we can infer that TRIM31 may be a valuable detective marker in primary CRC, and therapeutic intervention related to EMT suppression via TRIM31 can be a viable option for patients with advanced CRC.
Collapse
Affiliation(s)
- Haiyu Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University Shanghai, China
| | - Lu Yao
- Department of General Surgery, Zhongshan Hospital, Fudan University Shanghai, China
| | - Yuda Gong
- Department of General Surgery, Zhongshan Hospital, Fudan University Shanghai, China
| | - Bo Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University Shanghai, China
| |
Collapse
|
422
|
Vallée A, Lecarpentier Y. Crosstalk Between Peroxisome Proliferator-Activated Receptor Gamma and the Canonical WNT/β-Catenin Pathway in Chronic Inflammation and Oxidative Stress During Carcinogenesis. Front Immunol 2018; 9:745. [PMID: 29706964 PMCID: PMC5908886 DOI: 10.3389/fimmu.2018.00745] [Citation(s) in RCA: 264] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/26/2018] [Indexed: 12/19/2022] Open
Abstract
Inflammation and oxidative stress are common and co-substantial pathological processes accompanying, promoting, and even initiating numerous cancers. The canonical WNT/β-catenin pathway and peroxisome proliferator-activated receptor gamma (PPARγ) generally work in opposition. If one of them is upregulated, the other one is downregulated and vice versa. WNT/β-catenin signaling is upregulated in inflammatory processes and oxidative stress and in many cancers, although there are some exceptions for cancers. The opposite is observed with PPARγ, which is generally downregulated during inflammation and oxidative stress and in many cancers. This helps to explain in part the opposite and unidirectional profile of the canonical WNT/β-catenin signaling and PPARγ in these three frequent and morbid processes that potentiate each other and create a vicious circle. Many intracellular pathways commonly involved downstream will help maintain and amplify inflammation, oxidative stress, and cancer. Thus, many WNT/β-catenin target genes such as c-Myc, cyclin D1, and HIF-1α are involved in the development of cancers. Nuclear factor-kappaB (NFκB) can activate many inflammatory factors such as TNF-α, TGF-β, interleukin-6 (IL-6), IL-8, MMP, vascular endothelial growth factor, COX2, Bcl2, and inducible nitric oxide synthase. These factors are often associated with cancerous processes and may even promote them. Reactive oxygen species (ROS), generated by cellular alterations, stimulate the production of inflammatory factors such as NFκB, signal transducer and activator transcription, activator protein-1, and HIF-α. NFκB inhibits glycogen synthase kinase-3β (GSK-3β) and therefore activates the canonical WNT pathway. ROS activates the phosphatidylinositol 3 kinase/protein kinase B (PI3K/Akt) signaling in many cancers. PI3K/Akt also inhibits GSK-3β. Many gene mutations of the canonical WNT/β-catenin pathway giving rise to cancers have been reported (CTNNB1, AXIN, APC). Conversely, a significant reduction in the expression of PPARγ has been observed in many cancers. Moreover, PPARγ agonists promote cell cycle arrest, cell differentiation, and apoptosis and reduce inflammation, angiogenesis, oxidative stress, cell proliferation, invasion, and cell migration. All these complex and opposing interactions between the canonical WNT/β-catenin pathway and PPARγ appear to be fairly common in inflammation, oxidative stress, and cancers.
Collapse
Affiliation(s)
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| |
Collapse
|
423
|
Marzeda AM, Midwood KS. Internal Affairs: Tenascin-C as a Clinically Relevant, Endogenous Driver of Innate Immunity. J Histochem Cytochem 2018; 66:289-304. [PMID: 29385356 PMCID: PMC5958381 DOI: 10.1369/0022155418757443] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/29/2017] [Indexed: 12/20/2022] Open
Abstract
To protect against danger, the innate immune system must promptly and accurately sense alarm signals, and mount an appropriate response to restore homeostasis. One endogenous trigger of immunity is tenascin-C, a large hexameric protein of the extracellular matrix. Upregulated upon tissue injury and cellular stress, tenascin-C is expressed during inflammation and tissue remodeling, where it influences cellular behavior by interacting with a multitude of molecular targets, including other matrix components, cell surface proteins, and growth factors. Here, we discuss how these interactions confer upon tenascin-C distinct immunomodulatory capabilities that make this matrix molecule necessary for efficient tissue repair. We also highlight in vivo studies that provide insight into the consequences of misregulated tenascin-C expression on inflammation and fibrosis during a wide range of inflammatory diseases. Finally, we examine how its unique expression pattern and inflammatory actions make tenascin-C a viable target for clinical exploitation in both diagnostic and therapeutic arenas.
Collapse
Affiliation(s)
- Anna M Marzeda
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Kim S Midwood
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
424
|
Chhetri JK, de Souto Barreto P, Fougère B, Rolland Y, Vellas B, Cesari M. Chronic inflammation and sarcopenia: A regenerative cell therapy perspective. Exp Gerontol 2018; 103:115-123. [DOI: 10.1016/j.exger.2017.12.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/20/2017] [Accepted: 12/27/2017] [Indexed: 01/06/2023]
|
425
|
Pan X, Wu X, Yan D, Peng C, Rao C, Yan H. Acrylamide-induced oxidative stress and inflammatory response are alleviated by N-acetylcysteine in PC12 cells: Involvement of the crosstalk between Nrf2 and NF-κB pathways regulated by MAPKs. Toxicol Lett 2018; 288:55-64. [PMID: 29426002 DOI: 10.1016/j.toxlet.2018.02.002] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 01/04/2023]
Abstract
Acrylamide (ACR) is a classic neurotoxin in animals and humans. However, the mechanism underlying ACR neurotoxicity remains controversial, and effective prevention and treatment measures against this condition are scarce. This study focused on clarifying the crosstalk between the involved signaling pathways in ACR-induced oxidative stress and inflammatory response and investigating the protective effect of antioxidant N-acetylcysteine (NAC) against ACR in PC12 cells. Results revealed that ACR exposure led to oxidative stress characterized by significant increase in reactive oxygen species (ROS) and malondialdehyde (MDA) levels and glutathione (GSH) consumption. Inflammatory response was observed based on the dose-dependently increased levels of pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin 6 (IL-6). NAC attenuated ACR-induced enhancement of MDA and ROS levels and TNF-α generation. In addition, ACR activated nuclear transcription factor E2-related factor 2 (Nrf2) and nuclear factor-κB (NF-κB) signaling pathways. Knockdown of Nrf2 by siRNA significantly blocked the increased NF-κB p65 protein expression in ACR-treated PC12 cells. Down-regulation of NF-κB by specific inhibitor BAY11-7082 similarly reduced ACR-induced increase in Nrf2 protein expression. NAC treatment increased Nrf2 expression and suppressed NF-κB p65 expression to ameliorate oxidative stress and inflammatory response caused by ACR. Further results showed that mitogen-activated protein kinases (MAPKs) pathway was activated prior to the activation of Nrf2 and NF-κB pathways. Inhibition of MAPKs blocked Nrf2 and NF-κB pathways. Collectively, ACR activated Nrf2 and NF-κB pathways which were regulated by MAPKs. A crosstalk between Nrf2 and NF-κB pathways existed in ACR-induced cell damage. NAC protected against oxidative damage and inflammatory response induced by ACR by activating Nrf2 and inhibiting NF-κB pathways in PC12 cells.
Collapse
Affiliation(s)
- Xiaoqi Pan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China; School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xu Wu
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China
| | - Dandan Yan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China
| | - Cheng Peng
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Chaolong Rao
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Hong Yan
- Department of Health Toxicology, MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China.
| |
Collapse
|
426
|
Guo K, Hu L, Xi D, Zhao J, Liu J, Luo T, Ma Y, Lai W, Guo Z. PSRC1 overexpression attenuates atherosclerosis progression in apoE -/- mice by modulating cholesterol transportation and inflammation. J Mol Cell Cardiol 2018; 116:69-80. [PMID: 29378206 DOI: 10.1016/j.yjmcc.2018.01.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/16/2018] [Accepted: 01/20/2018] [Indexed: 01/13/2023]
Abstract
AIMS Human genome-wide association studies (GWAS) have found that proline/serine-rich coiled-coil 1 (PSRC1) encodes a protein that is associated with serum lipid levels and coronary artery disease. In addition, our previous study showed that the cholesterol efflux capacity is decreased in macrophages following a treatment silencing Psrc1, indicating that PSRC1 has anti-atherosclerotic effects. However, the role of PSRC1 in the development of atherosclerosis is unknown. This study aims to explore the effect of PSRC1 on atherosclerosis and its underlying mechanisms. METHOD AND RESULTS A recombinant adenovirus expressing Psrc1 (Ad-PSRC1) was constructed and transfected in RAW264.7 cells as well as injected intravenously into apoE-/- mice. The in vitro study showed that PSRC1 overexpression reduced the cellular cholesterol content, increased the cholesterol efflux capacity and inhibited foam cell formation by upregulating the expression of peroxisome proliferator-activated receptor γ (PPAR-γ) and liver X receptor α (LXR-α), which are key cholesterol transportation-related proteins. Infecting apoE-/- mice with Ad-PSRC1 inhibited the development of atherosclerotic lesions and enhanced atherosclerotic plaque stability. Consistent with these results, PSRC1 overexpression in apoE-/- mice decreased the plasma levels of TC, TG, LDL-C, TNF-α, IL-1β and IL-6, increased the plasma HDL-C levels and improved HDL function. Similarly, the PPAR-γ and LXR-α expression levels were upregulated in the liver and in peritoneal macrophages of PSRC1-overexpressing apoE-/- mice. Finally, the liver and peritoneal macrophages of apoE-/- mice displayed elevated expression of β-catenin, which is a direct downstream gene of PSRC1 and an upstream gene of PPAR-γ and LXR-α, but decreased activity of nuclear transcription factor (NF-κB), which acts as a key gene in the regulation of inflammation. CONCLUSIONS PSRC1 protects against the development of atherosclerosis and enhances the stability of plaques by modulating cholesterol transportation and inflammation in macrophages and the liver of apoE-/- mice.
Collapse
Affiliation(s)
- Kai Guo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Lu Hu
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Dan Xi
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jinzhen Zhao
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jichen Liu
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Tiantian Luo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Yusheng Ma
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Wenyan Lai
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Zhigang Guo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China.
| |
Collapse
|
427
|
Wu H, Jiang K, Guo S, Yang J, Zhao G, Qiu C, Deng G. IFN-τ Mediated Control of Bovine Major Histocompatibility Complex Class I Expression and Function via the Regulation of bta-miR-148b/152 in Bovine Endometrial Epithelial Cells. Front Immunol 2018; 9:167. [PMID: 29456541 PMCID: PMC5801426 DOI: 10.3389/fimmu.2018.00167] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 01/18/2018] [Indexed: 12/11/2022] Open
Abstract
IFN-τ, a type I interferon produced by the trophoblasts of ruminants, has various important immune functions, including effects on the expression of major histocompatibility complex (MHC) class I (MHC-I). A previous study has reported that IFN-τ promotes the expression of MHC-I molecules on endometrial cells. However, the immunological mechanisms by which IFN-τ regulates MHC-I molecules remain unknown. Here, we investigated which microRNA (miRNAs) may be involved in the regulation of MHC-I molecule expression and function in bovine endometrial epithelial cells (bEECs). By using TargetScan 6.2 and http://www.microRNA.org, two miRNAs were suggested to target the 3'UTR of the bovine MHC-I heavy chain: bta-miR-148b and bta-miR-152. Dual luciferase reporter and miRNA mimic/inhibitor assays suggested that bta-miR-148b/152 were negatively correlated with bovine MHC-I heavy chain genes. The function of the MHC-I heavy chain was then investigated using qRT-PCR, ELISA, western blotting, immunofluorescence, and RNA interference assays in primary bEECs and an endometrial epithelial cell line (BEND). The results demonstrated that bta-miR-148b/152 could promote TLR4-triggered inflammatory responses by targeting the bovine MHC-I heavy chain, and the MHC-I molecule negatively regulated TLR4-induced inflammatory reactions may through the Fps-SHP-2 pathway. Our discovery offers novel insight into negative regulation of the TLR4 pathway and elucidates the mechanism by which bovine MHC-I molecules control congenital inflammatory reactions.
Collapse
Affiliation(s)
- Haichong Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jing Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Changwei Qiu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
428
|
|
429
|
Regulation of influenza virus replication by Wnt/β-catenin signaling. PLoS One 2018; 13:e0191010. [PMID: 29324866 PMCID: PMC5764324 DOI: 10.1371/journal.pone.0191010] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/27/2017] [Indexed: 01/12/2023] Open
Abstract
Wnt/β-catenin signaling is an essential pathway in cell cycle control. Dysregulation of the Wnt/β-catenin signaling pathway during viral infection has been reported. In this study, we examined the effect of modulating Wnt/β-catenin signaling during influenza virus infection. The activation of the Wnt/β-catenin pathway by Wnt3a increased influenza virus mRNA and virus production in in vitro in mouse lung epithelial E10 cells and mRNA expresson of influenza virus genes in vivo in the lungs of mice infected with influenza virus A/Puerto Rico/8/34. However, the inhibition of Wnt/β-catenin signaling by iCRT14 reduced virus titer and viral gene expression in human lung epithelial A549 cells and viral replication in primary mouse alveolar epithelial cells infected with different influenza virus strains. Knockdown of β-catenin also reduced viral protein expression and virus production. iCRT14 acts at the early stage of virus replication. Treatment with iCRT14 inhibited the expression of the viral genes (vRNA, cRNA and mRNA) evaluated in this study. The intraperitoneal administration of iCRT14 reduced viral load, improved clinical signs, and partially protected mice from influenza virus infection.
Collapse
|
430
|
Rajagopal C, Lankadasari MB, Aranjani JM, Harikumar KB. Targeting oncogenic transcription factors by polyphenols: A novel approach for cancer therapy. Pharmacol Res 2018; 130:273-291. [PMID: 29305909 DOI: 10.1016/j.phrs.2017.12.034] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/30/2017] [Accepted: 12/31/2017] [Indexed: 02/06/2023]
Abstract
Inflammation is one of the major causative factor of cancer and chronic inflammation is involved in all the major steps of cancer initiation, progression metastasis and drug resistance. The molecular mechanism of inflammation driven cancer is the complex interplay between oncogenic and tumor suppressive transcription factors which include FOXM1, NF-kB, STAT3, Wnt/β- Catenin, HIF-1α, NRF2, androgen and estrogen receptors. Several products derived from natural sources modulate the expression and activity of multiple transcription factors in various tumor models as evident from studies conducted in cell lines, pre-clinical models and clinical samples. Further combination of these natural products along with currently approved cancer therapies added an additional advantage and they considered as promising targets for prevention and treatment of inflammation and cancer. In this review we discuss the application of multi-targeting natural products by analyzing the literature and future directions for their plausible applications in drug discovery.
Collapse
Affiliation(s)
- Chitra Rajagopal
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - Manendra Babu Lankadasari
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - Jesil Mathew Aranjani
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - K B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.
| |
Collapse
|
431
|
Huang L, Xiang M, Ye P, Zhou W, Chen M. Beta-catenin promotes macrophage-mediated acute inflammatory response after myocardial infarction. Immunol Cell Biol 2017; 96:100-113. [PMID: 29356094 DOI: 10.1111/imcb.1019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 10/01/2017] [Accepted: 10/02/2017] [Indexed: 01/01/2023]
Abstract
Regulatory mechanisms for acute inflammatory responses post myocardial infarction (MI) have yet to be fully understood. In particular, the mechanisms by which cardiac macrophages modulate MI-induced myocardial inflammation remains unclear. In this study, using a mouse MI model, we showed that β-catenin-mediated signaling was activated in cardiac macrophages post-MI, especially in Ly-6C-positive proinflammatory macrophages. Using a RAW264.7-based β-catenin reporter cell line, we confirmed the presence of active β-catenin and its downstream signaling in cardiac macrophages after MI. Moreover, lentivirus-mediated inducible expression of constitutively active β-catenin revealed that β-catenin plays a role in promoting the inflammatory response by RAW264.7 cells. Depletion of endogenous macrophages and adoptive transfer of active β-catenin-expressing RAW264.7 cells resulted in enhancement of acute myocardial inflammation in recipient mice after MI, as demonstrated by elevated levels of lymphocyte infiltrates and increased expression of proinflammatory cytokines. However, infarct volume, myocardial tissue repair, and left ventricle function were not influenced by the expression of active β-catenin in the adoptive transfer assay. Our research has demonstrated that β-catenin-mediated signaling is important for cardiac macrophages to modulate post-MI inflammatory responses. These findings may be valuable for developing novel therapeutic strategies for MI.
Collapse
Affiliation(s)
- Ling Huang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mei Xiang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Ye
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Zhou
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Manhua Chen
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
432
|
Magnesium Lithospermate B Suppresses Lipopolysaccharide-Induced Neuroinflammation in BV2 Microglial Cells and Attenuates Neurodegeneration in Lipopolysaccharide-Injected Mice. J Mol Neurosci 2017; 64:80-92. [PMID: 29196883 DOI: 10.1007/s12031-017-1007-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/24/2017] [Indexed: 12/11/2022]
Abstract
Chronic inflammation in the brain plays a critical role in major neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Microglia, the resident macrophages and intrinsic components of the central nervous system (CNS), appear to be the main effectors in this pathological process. Magnesium lithospermate B (MLB) is one of the major bioactive components of Radix Salviae miltiorrhizae, which has been documented to protect neurons against multiple types of neuronal injury. However, its functions on microglia and the related neuroinflammation remain unknown. In the present study, BV2 microglial cells were used to assess the anti-neuroinflammatory capacity of MLB. Our data show that treatment with MLB could not only suppress lipopolysaccharide (LPS)-induced proliferation and morphological changes, but also interfere with cell cycle progression in BV2 cells. More strikingly, it attenuated the production of the inflammatory mediator nitric oxide (NO) and a panel of pro-inflammatory cytokine in LPS-stimulated BV2 cells, including tumor necrosis factor-α (TNF-α), interleukin (IL)-1α, IL-1β, and IL-6, and also promoted a phenotypic switch from the M1 to the M2 phenotype. Additionally, an in vivo study showed that the administration of MLB could ameliorate lipopolysaccharide-induced neurodegeneration and microglial activation in the hippocampus of adult mice. Mechanistically, MLB blocked the activation of the NF-κB pathway upon LPS stimulation, indicating that the effects of MLB on microglia may be mediated by the NK-κB pathway. These results suggest the therapeutic potential of MLB as a novel anti-inflammatory and microglia-modulating drug for neurodegenerative diseases.
Collapse
|
433
|
Vallée A, Lecarpentier Y, Vallée JN. Thermodynamic Aspects and Reprogramming Cellular Energy Metabolism during the Fibrosis Process. Int J Mol Sci 2017; 18:ijms18122537. [PMID: 29186898 PMCID: PMC5751140 DOI: 10.3390/ijms18122537] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/10/2017] [Accepted: 11/21/2017] [Indexed: 01/06/2023] Open
Abstract
Fibrosis is characterized by fibroblast proliferation and fibroblast differentiation into myofibroblasts, which generate a relaxation-free contraction mechanism associated with excessive collagen synthesis in the extracellular matrix, which promotes irreversible tissue retraction evolving towards fibrosis. From a thermodynamic point of view, the mechanisms leading to fibrosis are irreversible processes that can occur through changing the entropy production rate. The thermodynamic behaviors of metabolic enzymes involved in fibrosis are modified by the dysregulation of both transforming growth factor β (TGF-β) signaling and the canonical WNT/β-catenin pathway, leading to aerobic glycolysis, called the Warburg effect. Molecular signaling pathways leading to fibrosis are considered dissipative structures that exchange energy or matter with their environment far from the thermodynamic equilibrium. The myofibroblastic cells arise from exergonic processes by switching the core metabolism from oxidative phosphorylation to glycolysis, which generates energy and reprograms cellular energy metabolism to induce the process of myofibroblast differentiation. Circadian rhythms are far-from-equilibrium thermodynamic processes. They directly participate in regulating the TGF-β and WNT/β-catenin pathways involved in energetic dysregulation and enabling fibrosis. The present review focusses on the thermodynamic implications of the reprogramming of cellular energy metabolism, leading to fibroblast differentiation into myofibroblasts through the positive interplay between TGF-β and WNT/β-catenin pathways underlying in fibrosis.
Collapse
Affiliation(s)
- Alexandre Vallée
- Laboratory of Mathematics and Applications (LMA), DACTIM, UMR CNRS 7348, CHU de Poitiers and University of Poitiers, 86021 Poitiers, France.
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), 77100 Meaux, France.
| | - Jean-Noël Vallée
- Laboratory of Mathematics and Applications (LMA), DACTIM, UMR CNRS 7348, CHU de Poitiers and University of Poitiers, 86021 Poitiers, France.
- CHU Amiens Picardie, University of Picardie Jules Verne (UPJV), 80025 Amiens, France.
| |
Collapse
|
434
|
Allen F, Bobanga ID, Rauhe P, Barkauskas D, Teich N, Tong C, Myers J, Huang AY. CCL3 augments tumor rejection and enhances CD8 + T cell infiltration through NK and CD103 + dendritic cell recruitment via IFNγ. Oncoimmunology 2017; 7:e1393598. [PMID: 29399390 PMCID: PMC5790335 DOI: 10.1080/2162402x.2017.1393598] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 01/08/2023] Open
Abstract
Inflammatory chemokines are critical contributors in attracting relevant immune cells to the tumor microenvironment and driving cellular interactions and molecular signaling cascades that dictate the ultimate outcome of host anti-tumor immune response. Therefore, rational application of chemokines in a spatial-temporal dependent manner may constitute an attractive adjuvant in immunotherapeutic approaches against cancer. Existing data suggest that the macrophage inflammatory protein (MIP)-1 family and related proteins, consisting of CCL3 (MIP-1α), CCL4 (MIP-1β), and CCL5 (RANTES), can be major determinant of immune cellular infiltration in certain tumors through their direct recruitment of antigen presenting cells, including dendritic cells (DCs) to the tumor site. In this study, we examined how CCL3 in a murine colon tumor microenvironment, CT26, enhances antitumor immunity. We identified natural killer (NK) cells as a major lymphocyte subtype that is preferentially recruited to the CCL3-rich tumor site. NK cells contribute to the overall IFNγ content, CD103+ DC accumulation, and augment the production of chemokines CXCL9 and CXCL10 for enhanced T cell recruitment. We further demonstrate that both soluble CCL3 and CCL3-secreting irradiated tumor vaccine can effectively halt the progression of established tumors in a spatial-dependent manner. Our finding implies an important contribution of NK in the CCL3 - CD103+ DC - CXCL9/10 signaling axis in determining tumor immune landscape within the tumor microenvironment.
Collapse
Affiliation(s)
- Frederick Allen
- Pathology, Case Western Reserve University School of Medicine, Wolstein Research Building, Cleveland, Ohio, United States
| | - Iuliana D. Bobanga
- Surgery, Case Western Reserve University School of Medicine, Wolstein Research Building, Cleveland, Ohio, United States
| | - Peter Rauhe
- Pediatrics, Case Western Reserve University School of Medicine, Wolstein Research Building, Cleveland, Ohio, United States
| | - Deborah Barkauskas
- Pediatrics, Case Western Reserve University School of Medicine, Wolstein Research Building, Cleveland, Ohio, United States
| | - Nathan Teich
- Pediatrics, Case Western Reserve University School of Medicine, Wolstein Research Building, Cleveland, Ohio, United States
| | - Caryn Tong
- Pediatrics, Case Western Reserve University School of Medicine, Wolstein Research Building, Cleveland, Ohio, United States
| | - Jay Myers
- Pediatrics, Case Western Reserve University School of Medicine, Wolstein Research Building, Cleveland, Ohio, United States
| | - Alex Y. Huang
- Pathology, Case Western Reserve University School of Medicine, Wolstein Research Building, Cleveland, Ohio, United States
- Pediatrics, Case Western Reserve University School of Medicine, Wolstein Research Building, Cleveland, Ohio, United States
- Angie Fowler AYA Cancer Institute, UH Rainbow Babies & Children's Hospital, Eulcid Avenue, Cleveland, Ohio, United States
| |
Collapse
|
435
|
Li F, Han F, Li H, Zhang J, Qiao X, Shi J, Yang L, Dong J, Luo M, Wei J, Liu X. Human placental mesenchymal stem cells of fetal origins-alleviated inflammation and fibrosis by attenuating MyD88 signaling in bleomycin-induced pulmonary fibrosis mice. Mol Immunol 2017; 90:11-21. [PMID: 28662409 DOI: 10.1016/j.molimm.2017.06.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 05/16/2017] [Accepted: 06/18/2017] [Indexed: 01/20/2023]
Abstract
Pulmonary fibrosis is a progressive lung disease that its pathogenic mechanism currently is incompletely understood. Toll-like receptor (TLR) signaling has recently been identified as a regulator of inflammation and pulmonary fibrosis. In addition, mesenchymal stem cells (MSCs) of different origins offer a great promise in treatment of idiopathic pulmonary fibrosis (IPF). However mechanisms of pathogenic roles of TLR signaling and therapeutic effects of MSCs in the IPF remain elusive. In present study, the involvement of TLR signaling and the therapeutic role of MSCs were interrogated in MyD88-deficient mice using human placental MSCs of fetal origins (hfPMSCs). The results showed an alleviated pulmonary inflammation and fibrosis in myeloid differentiation primary response gene 88 (MyD88)-deficient mice treated with bleomycin (BLM), accompanied with a reduced TGF-β signaling and production of pro-fibrotic cytokines, including TNF-α, IL-1β. An exposure of HLF1 lung fibroblasts, A549 epithelial cells and RAW264.7 macrophages to BLM led an increased expression of key components of MyD88 and TGF-β signaling cascades. Of interest, enforced expression and inhibition of MyD88 protein resulted in an enhanced and a reduced TGF-β signaling in above cells in the presence of BLM, respectively. However, the addition of TGF-β1 showed a marginally inhibitory effect on MyD88 signaling in these cells in the absence of BLM. Importantly, the administration of hfPMSCs could significantly attenuate BLM-induced pulmonary fibrosis in mice, along with a reduced hydroxyproline (HYP) deposition, MyD88 and TGF-β signaling activation, and production of pro-fibrotic cytokines. These results may suggest an importance of MyD88/TGF-β signaling axis in the tissue homeostasis and functional integrity of lung in response to injury, which may offer a novel target for treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Feng Li
- Center of Laboratory Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Department of Laboratory Medicine, College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Fei Han
- Department of Laboratory Medicine, College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Hui Li
- Department of Laboratory Medicine, College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Jia Zhang
- Department of Laboratory Medicine, College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Xia Qiao
- Center of Laboratory Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Juan Shi
- Center of Laboratory Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Li Yang
- The Center of Experimental Animals, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Jianda Dong
- Department of Pathology, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Meihui Luo
- Center of Laboratory Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Jun Wei
- Center of Laboratory Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Department of Laboratory Medicine, College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750004, China; Institute of Human Stem Cell Research, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Xiaoming Liu
- College of Life Science, Ningxia University, Yinchuan, Ningxia 750021, China.
| |
Collapse
|
436
|
Geng B, Zhang C, Wang C, Che Y, Mu X, Pan J, Xu C, Hu S, Yang J, Zhao T, Xu Y, Lv Y, Wen H, Liu Z, You Q. IκB-kinase-ε in the tumor microenvironment is essential for the progression of gastric cancer. Oncotarget 2017; 8:75298-75307. [PMID: 29088866 PMCID: PMC5650421 DOI: 10.18632/oncotarget.20778] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 07/30/2017] [Indexed: 01/26/2023] Open
Abstract
The tumor microenvironment is critical for tumor growth and metastasis, but the underlying molecular mechanisms are poorly understood. Recent studies have shown that IκB-kinase-ε (IKKε) is involved in the proliferation and migration of certain cancers. However, the functional role of IKKε in the progression of gastric cancer (GC) remains unknown. In this study, we found that high levels of IKKε expression in GC tumors were correlated with more advanced disease and poor overall survival of patients. Silencing of IKKε effectively suppressed the migratory and invasive capabilities of human GC cells in vitro and tumorigenicity and metastasis in vivo. Further analysis revealed that IKKε was also highly expressed in tumor-infiltrating lymphocytes. Moreover, it was involved in tumor-infiltrating T-cell-mediated invasion and metastasis. Knockdown of IKKε elevated T-cell antitumor immunity. These findings suggest that IKKε may be a novel prognostic marker and a potential therapeutic target in human GCs.
Collapse
Affiliation(s)
- Biao Geng
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Chen Zhang
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Chao Wang
- Department of Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Ying Che
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Xianmin Mu
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Jinshun Pan
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Che Xu
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Shi Hu
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Jing Yang
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Ting Zhao
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Yue Xu
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Yuanfang Lv
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Hao Wen
- Department of Surgery, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Zheng Liu
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
| | - Qiang You
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
- Key Laboratory for Aging & Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210011, China
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
437
|
Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. Interactions between TGF-β1, canonical WNT/β-catenin pathway and PPAR γ in radiation-induced fibrosis. Oncotarget 2017; 8:90579-90604. [PMID: 29163854 PMCID: PMC5685775 DOI: 10.18632/oncotarget.21234] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/17/2017] [Indexed: 12/16/2022] Open
Abstract
Radiation therapy induces DNA damage and inflammation leading to fibrosis. Fibrosis can occur 4 to 12 months after radiation therapy. This process worsens with time and years. Radiation-induced fibrosis is characterized by fibroblasts proliferation, myofibroblast differentiation, and synthesis of collagen, proteoglycans and extracellular matrix. Myofibroblasts are non-muscle cells that can contract and relax. Myofibroblasts evolve towards irreversible retraction during fibrosis process. In this review, we discussed the interplays between transforming growth factor-β1 (TGF-β1), canonical WNT/β-catenin pathway and peroxisome proliferator-activated receptor gamma (PPAR γ) in regulating the molecular mechanisms underlying the radiation-induced fibrosis, and the potential role of PPAR γ agonists. Overexpression of TGF-β and canonical WNT/β-catenin pathway stimulate fibroblasts accumulation and myofibroblast differentiation whereas PPAR γ expression decreases due to the opposite interplay of canonical WNT/β-catenin pathway. Both TGF-β1 and canonical WNT/β-catenin pathway stimulate each other through the Smad pathway and non-Smad pathways such as phosphatidylinositol 3-kinase/serine/threonine kinase (PI3K/Akt) signaling. WNT/β-catenin pathway and PPAR γ interact in an opposite manner. PPAR γ agonists decrease β-catenin levels through activation of inhibitors of the WNT pathway such as Smad7, glycogen synthase kinase-3 (GSK-3 β) and dickkopf-related protein 1 (DKK1). PPAR γ agonists also stimulate phosphatase and tensin homolog (PTEN) expression, which decreases both TGF-β1 and PI3K/Akt pathways. PPAR γ agonists by activating Smad7 decrease Smads pathway and then TGF-β signaling leading to decrease radiation-induced fibrosis. TGF-β1 and canonical WNT/β-catenin pathway promote radiation-induced fibrosis whereas PPAR γ agonists can prevent radiation-induced fibrosis.
Collapse
Affiliation(s)
- Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, Poitiers, France.,Laboratory of Mathematics and Applications (LMA), UMR CNRS 7348, University of Poitiers, Poitiers, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| | - Rémy Guillevin
- DACTIM, UMR CNRS 7348, University of Poitiers et CHU de Poitiers, Poitiers, France
| | - Jean-Noël Vallée
- Laboratory of Mathematics and Applications (LMA), UMR CNRS 7348, University of Poitiers, Poitiers, France.,CHU Amiens Picardie, University of Picardie Jules Verne (UPJV), Amiens, France
| |
Collapse
|
438
|
Zhou L, Dörfer CE, Chen L, Fawzy El-Sayed KM. Porphyromonas gingivalislipopolysaccharides affect gingival stem/progenitor cells attributes through NF-κB, but not Wnt/β-catenin, pathway. J Clin Periodontol 2017; 44:1112-1122. [DOI: 10.1111/jcpe.12777] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Lili Zhou
- Clinic of Conservative Dentistry and Periodontology; School of Dental Medicine; Christian-Albrechts Universität at Kiel; Kiel Germany
- Department of Oral Medicine; The Second Affiliated Hospital; School of Medicine; Zhejiang University; Hangzhou China
| | - Christof E. Dörfer
- Clinic of Conservative Dentistry and Periodontology; School of Dental Medicine; Christian-Albrechts Universität at Kiel; Kiel Germany
| | - Lili Chen
- Department of Oral Medicine; The Second Affiliated Hospital; School of Medicine; Zhejiang University; Hangzhou China
| | - Karim M. Fawzy El-Sayed
- Clinic of Conservative Dentistry and Periodontology; School of Dental Medicine; Christian-Albrechts Universität at Kiel; Kiel Germany
- Oral Medicine and Periodontology Department; Faculty of Oral and Dental Medicine; Cairo University; Cairo Egypt
| |
Collapse
|
439
|
Pei J, Fan L, Nan K, Li J, Shi Z, Dang X, Wang K. Excessive Activation of TLR4/NF-κB Interactively Suppresses the Canonical Wnt/β-catenin Pathway and Induces SANFH in SD Rats. Sci Rep 2017; 7:11928. [PMID: 28931847 PMCID: PMC5607349 DOI: 10.1038/s41598-017-12196-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/30/2017] [Indexed: 12/15/2022] Open
Abstract
Nuclear factor-kappa B (NF-κB) interactively affects the Wnt/β-catenin pathway and is closely related to different diseases. However, such crosstalk effect in steroid-associated necrosis of femoral head (SANFH) has not been fully explored and evaluated. In this study, early-stage SANFH was induced by two doses of lipopolysaccharide (LPS, 2 mg/kg/day) and three doses of methylprednisolone (MPS, 40 mg/kg/day). LPS and pyrrolidine dithiocarbamate (PDTC) were administered to activate the TLR4/NF-κB pathway and selectively block the activation of NF-κB, respectively. Results showed that PDTC treatment significantly reduced NF-κB expression, diminished inflammation, and effectively decreased bone resorption processes (osteoclastogenesis, adipogenesis, and apoptosis), which were evidently reinforced after osteonecrosis induction. Moreover, PDTC remarkably increased the interfered Wnt/β-catenin pathway and elevated bone formation processes (osteogenesis and angiogenesis). Ultimately, PDTC treatment effectively reduced the incidence of SANFH. Therefore, the excessive activation of TLR4/NF-κB may interactively suppress the Wnt/β-catenin pathway and induce SANFH. Hence, we propose NF-κB-targeted treatment as a novel therapeutic strategy for SANFH.
Collapse
Affiliation(s)
- Junpeng Pei
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an710004, Shaanxi Province, People's Republic of China
| | - Lihong Fan
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an710004, Shaanxi Province, People's Republic of China.
| | - Kai Nan
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an710004, Shaanxi Province, People's Republic of China
| | - Jia Li
- Department of Orthopaedics, First Affiliated Hospital of Xi'an Jiaotong University, School of Medicine, No. 277 Yanta Road, Xian, 710061, China
| | - Zhibin Shi
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an710004, Shaanxi Province, People's Republic of China
| | - Xiaoqian Dang
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an710004, Shaanxi Province, People's Republic of China
| | - Kunzheng Wang
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an710004, Shaanxi Province, People's Republic of China
| |
Collapse
|
440
|
Donson AM, Apps J, Griesinger AM, Amani V, Witt DA, Anderson RCE, Niazi TN, Grant G, Souweidane M, Johnston JM, Jackson EM, Kleinschmidt-DeMasters BK, Handler MH, Tan AC, Gore L, Virasami A, Gonzalez-Meljem JM, Jacques TS, Martinez-Barbera JP, Foreman NK, Hankinson TC. Molecular Analyses Reveal Inflammatory Mediators in the Solid Component and Cyst Fluid of Human Adamantinomatous Craniopharyngioma. J Neuropathol Exp Neurol 2017; 76:779-788. [PMID: 28859336 DOI: 10.1093/jnen/nlx061] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Pediatric adamantinomatous craniopharyngioma (ACP) is a highly solid and cystic tumor, often causing substantial damage to critical neuroendocrine structures such as the hypothalamus, pituitary gland, and optic apparatus. Paracrine signaling mechanisms driving tumor behavior have been hypothesized, with IL-6R overexpression identified as a potential therapeutic target. To identify potential novel therapies, we characterized inflammatory and immunomodulatory factors in ACP cyst fluid and solid tumor components. Cytometric bead analysis revealed a highly pro-inflammatory cytokine pattern in fluid from ACP compared to fluids from another cystic pediatric brain tumor, pilocytic astrocytoma. Cytokines and chemokines with particularly elevated concentrations in ACPs were IL-6, CXCL1 (GRO), CXCL8 (IL-8) and the immunosuppressive cytokine IL-10. These data were concordant with solid tumor compartment transcriptomic data from a larger cohort of ACPs, other pediatric brain tumors and normal brain. The majority of receptors for these cytokines and chemokines were also over-expressed in ACPs. In addition to IL-10, the established immunosuppressive factor IDO-1 was overexpressed by ACPs at the mRNA and protein levels. These data indicate that ACP cyst fluids and solid tumor components are characterized by an inflammatory cytokine and chemokine expression pattern. Further study regarding selective cytokine blockade may inform novel therapeutic interventions.
Collapse
Affiliation(s)
- Andrew M Donson
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - John Apps
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Andrea M Griesinger
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Vladimir Amani
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Davis A Witt
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Richard C E Anderson
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Toba N Niazi
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Gerald Grant
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Mark Souweidane
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - James M Johnston
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Eric M Jackson
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Bette K Kleinschmidt-DeMasters
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michael H Handler
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Aik-Choon Tan
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lia Gore
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Alex Virasami
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jose Mario Gonzalez-Meljem
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Thomas S Jacques
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Juan Pedro Martinez-Barbera
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Nicholas K Foreman
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Todd C Hankinson
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Developmental Biology and Cancer Programme, Great Ormond Street UCL Institute of Child Health, London, UK; Department of Neurological Surgery, Columbia University Medical Center, New York, New York; Division of Pediatric Neurosurgery, Department of Neurosurgery, Miami Children's Hospital, University of Miami/Miller School of Medicine, Miami, Florida; Department of Neurosurgery, Stanford University Medical Center, Palo Alto, California; Department of Neurological Surgery, Weill Medical College of Cornell University and Memorial Sloan-Kettering Cancer Center, New York, New York; Department of Neurosurgery, Children's Hospital Alabama, Birmingham, Alabama; Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pathology; Department of Neurosurgery; Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Histopathology, Great Ormond Street Hospital, NHS Trust, London, UK; Morgan Adams Foundation Pediatric Brain Tumor Research Program; Pediatric Neurosurgery, Children's Hospital Colorado; and Adult and Child Center for Health Outcomes Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | |
Collapse
|
441
|
Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. PPARγ agonists: Potential treatments for exudative age-related macular degeneration. Life Sci 2017; 188:123-130. [PMID: 28887057 DOI: 10.1016/j.lfs.2017.09.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/29/2017] [Accepted: 09/05/2017] [Indexed: 12/22/2022]
Abstract
Choroidal neovascularization (CNV) characterizes the progression of exudative age-related macular degeneration (AMD) with the deterioration in the central vision. Vascular inflammation, and overproduction of inflammatory cytokines, growth factors and aberrant endothelial cell migration, initiate defective blood vessel proliferation in exudative AMD. CNV formation is initiated by the interplay between inflammation, the hallmark of exudative AMD, and the activation of WNT/β-catenin pathway. Upregulation of WNT/β-catenin pathway involves activation of PI3K/Akt pathway and then the Warburg effect to produce lactate. Lactate production generates VEGF expression and then participates to the initiation of CNV in exudative AMD. WNT/β-catenin pathway and PPARγ act in an opposite manner in several diseases. We focus this review on the interplay between PPARγ and canonical WNT/β-catenin pathway and the anti-inflammatory role of PPARγ in exudative AMD. In exudative AMD, PPARγ agonists downregulate inflammation and the WNT/β-catenin pathway. PPARγ agonists can appear as promising treatment against the initiation and the progression of CNV in exudative AMD.
Collapse
Affiliation(s)
- Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, Poitiers, France; Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, France.
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| | - Rémy Guillevin
- Université de Poitiers et CHU de Poitiers, DACTIM, Laboratoire de Mathématiques et Applications, UMR CNRS 7348, SP2MI, Futuroscope, France
| | - Jean-Noël Vallée
- Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, France; CHU Amiens Picardie, Université Picardie Jules Verne (UPJV), Amiens, France
| |
Collapse
|
442
|
Torre E. Molecular signaling mechanisms behind polyphenol-induced bone anabolism. PHYTOCHEMISTRY REVIEWS : PROCEEDINGS OF THE PHYTOCHEMICAL SOCIETY OF EUROPE 2017; 16:1183-1226. [PMID: 29200988 PMCID: PMC5696504 DOI: 10.1007/s11101-017-9529-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 08/20/2017] [Indexed: 05/08/2023]
Abstract
For millennia, in the different cultures all over the world, plants have been extensively used as a source of therapeutic agents with wide-ranging medicinal applications, thus becoming part of a rational clinical and pharmacological investigation over the years. As bioactive molecules, plant-derived polyphenols have been demonstrated to exert many effects on human health by acting on different biological systems, thus their therapeutic potential would represent a novel approach on which natural product-based drug discovery and development could be based in the future. Many reports have provided evidence for the benefits derived from the dietary supplementation of polyphenols in the prevention and treatment of osteoporosis. Polyphenols are able to protect the bone, thanks to their antioxidant properties, as well as their anti-inflammatory actions by involving diverse signaling pathways, thus leading to bone anabolic effects and decreased bone resorption. This review is meant to summarize the research works performed so far, by elucidating the molecular mechanisms of action of polyphenols in a bone regeneration context, aiming at a better understanding of a possible application in the development of medical devices for bone tissue regeneration.
Collapse
Affiliation(s)
- Elisa Torre
- Nobil Bio Ricerche srl, Via Valcastellana, 26, 14037 Portacomaro, AT Italy
| |
Collapse
|
443
|
Suppression of Wnt Signaling and Osteogenic Changes in Vascular Smooth Muscle Cells by Eicosapentaenoic Acid. Nutrients 2017; 9:nu9080858. [PMID: 28796175 PMCID: PMC5579651 DOI: 10.3390/nu9080858] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 01/01/2023] Open
Abstract
Vascular medial calcification is often observed in patients with arteriosclerosis. It is also associated with systolic hypertension, wide pulse pressure, and fluctuation of blood pressure, which results in cardiovascular events. Eicosapentaenoic acid (EPA) has been shown to suppress vascular calcification in previous animal experiments. We investigated the inhibitory effects of EPA on Wnt signaling, which is one of the important signaling pathways involved in vascular calcification. Intake of food containing 5% EPA resulted in upregulation of the mRNA expression of Klotho, an intrinsic inhibitor of Wnt signaling, in the kidneys of wild-type mice. Expression levels of β-catenin, an intracellular signal transducer in the Wnt signaling pathway, were increased in the aortas of Klotho mutant (kl/kl) mice compared to the levels in the aortas of wild-type mice. Wnt3a or BIO, a GSK-3 inhibitor that activates β-catenin signaling, upregulated mRNA levels of AXIN2 and LEF1, Wnt signaling marker genes, and RUNX2 and BMP4, early osteogenic genes, in human aorta smooth muscle cells. EPA suppressed the upregulation of AXIN2 and BMP4. The effect of EPA was cancelled by T0070907, a PPARγ inhibitor. The results suggested that EPA could suppress vascular calcification via the inhibition of Wnt signaling in osteogenic vascular smooth muscle cells via PPARγ activation.
Collapse
|
444
|
Koch S. Extrinsic control of Wnt signaling in the intestine. Differentiation 2017; 97:1-8. [PMID: 28802143 DOI: 10.1016/j.diff.2017.08.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/28/2017] [Accepted: 08/04/2017] [Indexed: 12/15/2022]
Abstract
The canonical Wnt/β-catenin signaling pathway is a central regulator of development and tissue homeostasis. In the intestine, Wnt signaling is primarily known as the principal organizer of epithelial stem cell identity and proliferation. Within the last decade, numerous scientific breakthroughs have shed light on epithelial self-organization in the gut, and organoids are now routinely used to study stem cell biology and intestinal pathophysiology. The contribution of non-epithelial cells to Wnt signaling in the gut has received less attention. However, there is mounting evidence that stromal cells are a rich source of Wnt pathway activators and inhibitors, which can dynamically shape Wnt signaling to control epithelial proliferation and restitution. Elucidating the extent and mechanisms of paracrine Wnt signaling in the intestine has the potential to broaden our understanding of epithelial homeostasis, and may be of particular relevance for disorders such as inflammatory bowel diseases and colitis-associated cancers.
Collapse
Affiliation(s)
- Stefan Koch
- Department of Clinical and Experimental Medicine (IKE), Faculty of Health Sciences, Linköping University, S-581 85 Linköping, Sweden; Wallenberg Centre for Molecular Medicine (WCMM), Linköping University, S-581 85 Linköping, Sweden.
| |
Collapse
|
445
|
Yang HF, Yu M, Jin HD, Yao JQ, Lu ZL, Yabasin IB, Yan Q, Wen QP. Fentanyl Promotes Breast Cancer Cell Stemness and Epithelial-Mesenchymal Transition by Upregulating α1, 6-Fucosylation via Wnt/β-Catenin Signaling Pathway. Front Physiol 2017; 8:510. [PMID: 28798691 PMCID: PMC5526971 DOI: 10.3389/fphys.2017.00510] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/04/2017] [Indexed: 01/14/2023] Open
Abstract
Cancer pain is a common and severe complication of human breast cancer, and relieving pain is fundamental strategy in the treatment. Fentanyl, as an opioid analgesic, is widely used in breast cancer patients. However, little is known about its effects on stemness and epithelial-mesenchymal transition (EMT) of breast cancer cells. Aberrant protein glycosylation is involved in cancer malignancy. The α1, 6-fucosylation is an important type of glycosylation, and the elevated α1, 6-fucosylation catalyzed by fucosyltransferase VIII (FUT8) is found in many tumors. However, whether 1, 6-fucosylation is involved in regulating stemness and EMT, and stimulated by fentanyl is not clear. In this study, we found that fentanyl induced stemness and EMT in MCF-7 and MDA-MB-231 breast cancer cells by analysis of sphere formation, expression of stemness markers (Sox2, Oct4) and EMT markers (N-cadherin, E-cadherin and Vimentin). Results also showed that fentanyl upregulated FUT8 gene and protein expression by qPCR, Western blot and immunofluorescent staining, as well as α1, 6-fucosylation level by Lectin blot and Lectin fluorescent staining. Furthermore, decreased or blocked α1, 6-fucosylation by FUT8 siRNA transfection or LCA Lectin blockage reduced stemness and EMT. Additionally, fentanyl activated the key molecules and target genes in Wnt/β-catenin signaling pathway. LGK-974 (an inhibitor of Wnt ligands) suppressed fentanyl-mediated upregulation of α1, 6-fucosylation, stemness and EMT. The results of tumor xenograft demonstrated that fentanyl enhanced tumor growth, α1, 6-fucosylation, stemness and EMT. Taken together, our study reveals that fentanyl upregulated FUT8 expression, which increased α1, 6-fucosylation level through activation of Wnt/β-catenin signaling pathway, thereby, induce stemness and EMT of breast cancer cells. This study suggest a potential side effect of fentanyl in the treatment of cancer, which may guide the safety of fentanyl in the clinical application.
Collapse
Affiliation(s)
- Hong-Fang Yang
- Department of Anesthesiology, First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Ming Yu
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Department of Biochemistry and Molecular Biology, Dalian Medical UniversityDalian, China
| | - Hui-Dan Jin
- Department of Anesthesiology, First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Jia-Qi Yao
- Department of Anesthesiology, Affiliated Xinhua Hospital of Dalian UniversityDalian, China
| | - Zhi-Li Lu
- Department of Anesthesiology, First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Iddrisu B Yabasin
- Department of Anesthesiology, First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Qiu Yan
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Department of Biochemistry and Molecular Biology, Dalian Medical UniversityDalian, China
| | - Qing-Ping Wen
- Department of Anesthesiology, First Affiliated Hospital of Dalian Medical UniversityDalian, China
| |
Collapse
|
446
|
WNT ligands contribute to the immune response during septic shock and amplify endotoxemia-driven inflammation in mice. Blood Adv 2017; 1:1274-1286. [PMID: 29296769 DOI: 10.1182/bloodadvances.2017006163] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/07/2017] [Indexed: 12/12/2022] Open
Abstract
Improved understanding of the molecular mechanisms underlying dysregulated inflammatory responses in severe infection and septic shock is urgently needed to improve patient management and identify new therapeutic opportunities. The WNT signaling pathway has been implicated as a novel constituent of the immune response to infection, but its contribution to the host response in septic shock is unknown. Although individual WNT proteins have been ascribed pro- or anti-inflammatory functions, their concerted contributions to inflammation in vivo remain to be clearly defined. Here we report differential expression of multiple WNT ligands in whole blood of patients with septic shock and reveal significant correlations with inflammatory cytokines. Systemic challenge of mice with lipopolysaccharide (LPS) similarly elicited differential expression of multiple WNT ligands with correlations between WNT and cytokine expression that partially overlap with the findings in human blood. Molecular regulators of WNT expression during microbial encounter in vivo are largely unexplored. Analyses in gene-deficient mice revealed differential contributions of Toll-like receptor signaling adaptors, a positive role for tumor necrosis factor, but a negative regulatory role for interleukin (IL)-12/23p40 in the LPS-induced expression of Wnt5b, Wnt10a, Wnt10b, and Wnt11. Pharmacologic targeting of bottlenecks of the WNT network, WNT acylation and β-catenin activity, diminished IL-6, tumor necrosis factor, and IL-12/23p40 in serum of LPS-challenged mice and cultured splenocytes, whereas IL-10 production remained largely unaffected. Taken together, our data support the conclusion that the concerted action of WNT proteins during severe infection and septic shock promotes inflammation, and that this is, at least in part, mediated by WNT/β-catenin signaling.
Collapse
|
447
|
Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. Aerobic Glycolysis Hypothesis Through WNT/Beta-Catenin Pathway in Exudative Age-Related Macular Degeneration. J Mol Neurosci 2017; 62:368-379. [PMID: 28689265 DOI: 10.1007/s12031-017-0947-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 06/28/2017] [Indexed: 12/18/2022]
Abstract
Exudative age-related macular degeneration (AMD) is characterized by molecular mechanisms responsible for the initiation of choroidal neovascularization (CNV). Inflammatory processes are associated with upregulation of the canonical WNT/beta-catenin pathway in exudative AMD. We focus this review on the link between WNT/beta-catenin pathway activation and neovascular progression in exudative AMD through activation of aerobic glycolysis for production of angiogenic factors. Increased WNT/beta-catenin pathway involves hexokinase 2 (HK2) and pyruvate kinase M2 (PKM2). WNT/beta-catenin pathway stimulates PI3K/Akt pathway and then HIF-1alpha which activates glycolytic enzymes: glucose transporter (Glut), pyruvate dehydrogenase kinase 1 (PDK1), lactate dehydrogenase A (LDH-A), and monocarboxylate lactate transporter (MCT-1). This phenomenon is called aerobic glycolysis or the Warburg effect. Consequently, phosphorylation of PDK-1 inhibits the pyruvate dehydrogenase complex (PDH). Thus, a large part of pyruvate cannot be converted into acetyl-CoA in mitochondria and only a part of acetyl-CoA can enter the tricarboxylic acid cycle. Cytosolic pyruvate is converted into lactate through the action of LDH-A. In exudative AMD, high level of cytosolic lactate is correlated with increase of VEGF expression, the angiogenic factor of CNV. Photoreceptors in retina cells can metabolize glucose through aerobic glycolysis to protect them against oxidative damage, as cancer cells do.
Collapse
Affiliation(s)
- Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, Poitiers, France.
- Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, 11 Boulevard Marie et Pierre Curie, Poitiers, France.
| | | | - Rémy Guillevin
- DACTIM, Laboratoire de Mathématiques et Applications, Université de Poitiers et CHU de Poitiers, UMR CNRS 7348, SP2MI Futuroscope, Chasseneuil-du-Poitou, France
| | - Jean-Noël Vallée
- Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, 11 Boulevard Marie et Pierre Curie, Poitiers, France
- CHU Amiens Picardie, Université Picardie Jules Verne (UPJV), Amiens, France
| |
Collapse
|
448
|
Distinct Roles of Wnt/ β-Catenin Signaling in the Pathogenesis of Chronic Obstructive Pulmonary Disease and Idiopathic Pulmonary Fibrosis. Mediators Inflamm 2017; 2017:3520581. [PMID: 28588349 PMCID: PMC5447271 DOI: 10.1155/2017/3520581] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/29/2017] [Accepted: 04/12/2017] [Indexed: 02/07/2023] Open
Abstract
Wnt signaling pathways are tightly controlled under a physiological condition, under which they play key roles in many biological functions, including cell fate specification and tissue regeneration. Increasing lines of evidence recently demonstrated that a dysregulated activation of Wnt signaling, particularly the Wnt/β-catenin signaling, was involved in the pathogenesis of chronic pulmonary diseases, such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). In this respect, Wnt signaling interacts with other cellular signaling pathways to regulate the initiation and pathogenic procedures of airway inflammation and remodeling, pulmonary myofibroblast proliferation, epithelial-to-mesenchymal transition (EMT), and development of emphysema. Intriguingly, Wnt/β-catenin signaling is activated in IPF; an inhibition of this signaling leads to an alleviation of pulmonary inflammation and fibrosis in experimental models. Conversely, Wnt/β-catenin signaling is inactivated in COPD tissues, and its reactivation results in an amelioration of airspace enlargement with a restored alveolar epithelial structure and function in emphysema models. These studies thus imply distinct mechanisms of Wnt/β-catenin signaling in the pathogenesis of these two chronic pulmonary diseases, indicating potential targets for COPD and IPF treatments. This review article aims to summarize the involvement and pathogenic roles of Wnt signaling pathways in the COPD and IPF, with a focus on the implication of Wnt/β-catenin signaling as underlying mechanisms and therapeutic targets in these two incurable diseases.
Collapse
|
449
|
Archer TC, Mahoney EL, Pomeroy SL. Medulloblastoma: Molecular Classification-Based Personal Therapeutics. Neurotherapeutics 2017; 14:265-273. [PMID: 28386677 PMCID: PMC5398996 DOI: 10.1007/s13311-017-0526-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Recent advances in cancer genomics have revealed 4 distinct subgroups of medulloblastomas, each with unique transcription profiles, DNA alterations and clinical outcome. Molecular classification of medulloblastomas improves predictions of clinical outcome, allowing more accurate matching of intensity of conventional treatments with chemotherapy and radiation to overall prognosis and setting the stage for the introduction of targeted therapies.
Collapse
Affiliation(s)
- Tenley C Archer
- Department of Neurology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | | | - Scott L Pomeroy
- Department of Neurology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
450
|
Zolezzi JM, Inestrosa NC. Wnt/TLR Dialog in Neuroinflammation, Relevance in Alzheimer's Disease. Front Immunol 2017; 8:187. [PMID: 28286503 PMCID: PMC5323396 DOI: 10.3389/fimmu.2017.00187] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/09/2017] [Indexed: 12/22/2022] Open
Abstract
The innate immune system (IIS) represents the first line of defense against exogenous and endogenous harmful stimuli. Different types of pathogens and diverse molecules can activate the IIS via a ligand-receptor mechanism. Cytokine release, recruitment of immunocompetent cells, and inflammation constitute the initial steps in an IIS-mediated response. While balanced IIS activity can resolve a harmful event, an altered response, such as deficient or persistent IIS activity, will have a critical effect on organism homeostasis. In this regard, chronic IIS activation has been associated with a wide range of diseases, including chronic inflammatory disorders (inflammatory bowel disease, arthritis, chronic obstructive pulmonary disease, among others), cancer and, more recently, neurodegenerative disorders. The relevance of the immune response, particularly inflammation, in the context of neurodegeneration has motivated rigorous research focused on unveiling the mechanisms underlying this response. Knowledge regarding the molecular hallmarks of the innate immune response and understanding signaling pathway cross talk are critical for developing new therapeutic strategies aimed at modulating the neuroinflammatory response within the brain. In the present review, we discuss the IIS in the central nervous system, particularly the cross talk between the toll-like receptor-signaling cascade and the wingless-related MMTV integration site (Wnt) signaling pathway and its relevance in neurodegenerative disorders such as Alzheimer's disease.
Collapse
Affiliation(s)
- Juan M Zolezzi
- Centro de Envejecimiento y Regeneración (CARE-UC), Facultad de Ciencias Biológicas, Departamento de Biología Celular y Molecular, P. Universidad Católica de Chile , Santiago , Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE-UC), Facultad de Ciencias Biológicas, Departamento de Biología Celular y Molecular, P. Universidad Católica de Chile, Santiago, Chile; Centre for Healthy Brain Ageing, Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|