51
|
Tran NQV, Nguyen AN, Takabe K, Yamagata Z, Miyake K. Pre-treatment with amitriptyline causes epigenetic up-regulation of neuroprotection-associated genes and has anti-apoptotic effects in mouse neuronal cells. Neurotoxicol Teratol 2017; 62:1-12. [PMID: 28511916 DOI: 10.1016/j.ntt.2017.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 05/09/2017] [Accepted: 05/11/2017] [Indexed: 12/12/2022]
Abstract
Antidepressants, such as imipramine and fluoxetine, are known to alter gene expression patterns by inducing changes in the epigenetic status of neuronal cells. There is also some evidence for the anti-apoptotic effect of various groups of antidepressants; however, this effect is complicated and cell-type dependent. Antidepressants of the tricyclic group, in particular amitriptyline, have been suggested to be beneficial in the treatment of neurodegenerative disorders. We examined whether amitriptyline exerts an anti-apoptotic effect via epigenetic mechanisms. Using DNA microarray, we analyzed global gene expression in mouse primary cultured neocortical neurons after treatment with amitriptyline and imipramine. The neuroprotection-associated genes, activating transcription factor 3 (Atf3) and heme oxygenase 1 (Hmox1), were up-regulated at both mRNA and protein levels by treatment with amitriptyline. Quantitative chromatin immunoprecipitation assay revealed that amitriptyline increased enrichments of trimethylation of histone H3 lysine 4 in the promoter regions of Atf3 and Hmox1 and acetylation of histone H3 lysine 9 in the promoter regions of Atf3, which indicate an active epigenetic status. Amitriptyline pre-treatment attenuated 1-methyl-4-phenylpyridinium ion (MPP+)- or amyloid β peptide 1-42 (Aβ1-42)-induced neuronal cell death and inhibited the activation of extracellular signal-regulated kinase 1 and 2 (ERK1/2). We found that Atf3 and Hmox1 were also up-regulated after Aβ1-42 treatment, and were further increased when pre-treated with amitriptyline. Interestingly, the highest up-regulation of Atf3 and Hmox1, at least at mRNA level, was observed after co-treatment with Aβ1-42 and amitriptyline, together with the loss of the neuroprotective effect. These findings suggest preconditioning and neuroprotective effects of amitriptyline; however, further investigations are needed for clarifying the contribution of epigenetic up-regulation of Atf3 and Hmox1 genes.
Collapse
Affiliation(s)
- Nguyen Quoc Vuong Tran
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - An Nghia Nguyen
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Kyoko Takabe
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Zentaro Yamagata
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan
| | - Kunio Miyake
- Department of Health Sciences, Graduate School of Interdisciplinary Research, University of Yamanashi, 1110, Shimokato, Chuo, Yamanashi 409-3898, Japan.
| |
Collapse
|
52
|
Ban N, Siegfried CJ, Lin JB, Shui YB, Sein J, Pita-Thomas W, Sene A, Santeford A, Gordon M, Lamb R, Dong Z, Kelly SC, Cavalli V, Yoshino J, Apte RS. GDF15 is elevated in mice following retinal ganglion cell death and in glaucoma patients. JCI Insight 2017; 2:91455. [PMID: 28469085 DOI: 10.1172/jci.insight.91455] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 04/04/2017] [Indexed: 10/19/2022] Open
Abstract
Glaucoma is the second leading cause of blindness worldwide. Physicians often use surrogate endpoints to monitor the progression of glaucomatous neurodegeneration. These approaches are limited in their ability to quantify disease severity and progression due to inherent subjectivity, unreliability, and limitations of normative databases. Therefore, there is a critical need to identify specific molecular markers that predict or measure glaucomatous neurodegeneration. Here, we demonstrate that growth differentiation factor 15 (GDF15) is associated with retinal ganglion cell death. Gdf15 expression in the retina is specifically increased after acute injury to retinal ganglion cell axons and in a murine chronic glaucoma model. We also demonstrate that the ganglion cell layer may be one of the sources of secreted GDF15 and that GDF15 diffuses to and can be detected in aqueous humor (AH). In validating these findings in human patients with glaucoma, we find not only that GDF15 is increased in AH of patients with primary open angle glaucoma (POAG), but also that elevated GDF15 levels are significantly associated with worse functional outcomes in glaucoma patients, as measured by visual field testing. Thus, GDF15 maybe a reliable metric of glaucomatous neurodegeneration, although further prospective validation studies will be necessary to determine if GDF15 can be used in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Rajendra S Apte
- Departments of Ophthalmology.,Medicine, and.,Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
53
|
Low JK, Ambikairajah A, Shang K, Brown DA, Tsai VWW, Breit SN, Karl T. First Behavioural Characterisation of a Knockout Mouse Model for the Transforming Growth Factor (TGF)-β Superfamily Cytokine, MIC-1/GDF15. PLoS One 2017; 12:e0168416. [PMID: 28081177 PMCID: PMC5231335 DOI: 10.1371/journal.pone.0168416] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 11/30/2016] [Indexed: 11/28/2022] Open
Abstract
Macrophage inhibitory cytokine-1 (MIC-1), also known as growth differentiation factor 15 (GDF15), is a stress response cytokine. MIC-1/GDF15 is secreted into the cerebrospinal fluid and increased levels of MIC-1/GDF15 are associated with a variety of diseases including cognitive decline. Furthermore, Mic-1/Gdf15 knockout mice (Mic-1 KO) weigh more, have increased adiposity, associated with increased spontaneous food intake, and exhibit reduced basal energy expenditure and physical activity. The current study was designed to comprehensively determine the role of MIC-1/GDF15 on behavioural domains of male and female knockout mice including locomotion, exploration, anxiety, cognition, social behaviours, and sensorimotor gating. Mic-1 KO mice exhibited a task-dependent increase in locomotion and exploration and reduced anxiety-related behaviours across tests. Spatial working memory and social behaviours were not affected by Mic-1/Gdf15 deficiency. Interestingly, knockout mice formed an increased association with the conditioned stimulus in fear conditioning testing and also displayed significantly improved prepulse inhibition. Overall sex effects were evident for social behaviours, fear conditioning, and sensorimotor gating. This is the first study defining the role of Mic-1/Gdf15 in a number of behavioural domains. Whether the observed impact is based on direct actions of Mic-1/Gdf15 deficiency on the CNS or whether the behavioural effects are mediated by indirect actions on e.g. other neurotransmitter systems must be clarified in future studies.
Collapse
Affiliation(s)
- Jac Kee Low
- Neuroscience Research Australia (NeuRA), Randwick, New South Wales, Australia
| | | | - Kani Shang
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - David A. Brown
- St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Darlinghurst, New South Wales, Australia
- Westmead Institute for Medical Research, The Institute for Clinical Pathology and Medical Research and Westmead Hospital, Westmead, New South Wales, Australia
| | - Vicky W. W. Tsai
- St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Darlinghurst, New South Wales, Australia
| | - Samuel N. Breit
- St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Darlinghurst, New South Wales, Australia
| | - Tim Karl
- Neuroscience Research Australia (NeuRA), Randwick, New South Wales, Australia
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
- * E-mail:
| |
Collapse
|
54
|
Tedeschi A, Omura T, Costigan M. CNS repair and axon regeneration: Using genetic variation to determine mechanisms. Exp Neurol 2017; 287:409-422. [PMID: 27163547 PMCID: PMC5097896 DOI: 10.1016/j.expneurol.2016.05.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/02/2016] [Accepted: 05/05/2016] [Indexed: 10/21/2022]
Abstract
The importance of genetic diversity in biological investigation has been recognized since the pioneering studies of Gregor Johann Mendel and Charles Darwin. Research in this area has been greatly informed recently by the publication of genomes from multiple species. Genes regulate and create every part and process in a living organism, react with the environment to create each living form and morph and mutate to determine the history and future of each species. The regenerative capacity of neurons differs profoundly between animal lineages and within the mammalian central and peripheral nervous systems. Here, we discuss research that suggests that genetic background contributes to the ability of injured axons to regenerate in the mammalian central nervous system (CNS), by controlling the regulation of specific signaling cascades. We detail the methods used to identify these pathways, which include among others Activin signaling and other TGF-β superfamily members. We discuss the potential of altering these pathways in patients with CNS damage and outline strategies to promote regeneration and repair by combinatorial manipulation of neuron-intrinsic and extrinsic determinants.
Collapse
Affiliation(s)
- Andrea Tedeschi
- German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany.
| | - Takao Omura
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Michael Costigan
- FM Kirby Neurobiology Center and Anesthesia Department, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
55
|
Chai YL, Hilal S, Chong JP, Ng YX, Liew OW, Xu X, Ikram MK, Venketasubramanian N, Richards AM, Lai MK, Chen CP. Growth differentiation factor-15 and white matter hyperintensities in cognitive impairment and dementia. Medicine (Baltimore) 2016; 95:e4566. [PMID: 27537582 PMCID: PMC5370808 DOI: 10.1097/md.0000000000004566] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Vascular pathology plays an important role in the development of cognitive decline and dementia. In this context, growth differentiation factor-15 (GDF-15) has been suggested to be a biomarker due to its regulatory roles in inflammatory and trophic responses during tissue injury. However, limited data exist on the associations of GDF-15 with either cerebrovascular disease (CeVD) burden or the spectrum of cognitive impairment. Therefore, we aimed to study peripheral levels of GDF-15 incognitive impairment no dementia (CIND) or Alzheimer disease (AD) subjects assessed for CeVD using a case-control cohort design, with cases recruited from memory clinics and controls from memory clinics and the community. All subjects underwent detailed neuropsychological assessment, 3-Tesla magnetic resonance imaging, and venous blood draw. Subjects were classified as CIND or AD based on clinical criteria, while significant CeVD was defined as the presence of cortical infarcts and/or 2 lacunes or more, and/or confluent white matter hyperintensities (WMHs) in 2 or more brain regions. A total of 324 subjects were included in the study, of whom 80 had no cognitive impairment, 144 CIND and 100with AD. Higher GDF-15 levels were significantly associated with disease groups, especially in the presence of CeVD, namely, CIND with CeVD (odds ratios [OR]: 7.21; 95% confidence interval [CI]: 2.14-24.27) and AD with CeVD (OR: 21.87; 95% CI: 2.01-237.43). Among the different CeVD markers, only WMH was associated with higher GDF-15 levels (OR: 3.97; 95% CI: 1.79-8.83). The associations between GDF-15 and cognitive impairment as well as with WMH remained significant after excluding subjects with cardiovascular diseases. In conclusion, we showed that increased GDF-15 may be a biomarker for CIND and AD in subjects with WMH.
Collapse
Affiliation(s)
- Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore
- Memory Aging and Cognition Centre, National University Health System
| | - Saima Hilal
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore
- Memory Aging and Cognition Centre, National University Health System
| | - Jenny P.C. Chong
- Cardiovascular Research Institute, National University Heart Centre, Singapore
| | - Yan Xia Ng
- Cardiovascular Research Institute, National University Heart Centre, Singapore
| | - Oi Wah Liew
- Cardiovascular Research Institute, National University Heart Centre, Singapore
| | - Xin Xu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore
- Memory Aging and Cognition Centre, National University Health System
| | - Mohammad Kamran Ikram
- Memory Aging and Cognition Centre, National University Health System
- Departments of Neurology and Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - A. Mark Richards
- Cardiovascular Research Institute, National University Heart Centre, Singapore
| | - Mitchell K.P. Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore
- Memory Aging and Cognition Centre, National University Health System
- Correspondence: Mitchell K.P. Lai, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09–01, 14 Medical Drive, 117599 Singapore (e-mail: )
| | - Christopher P. Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore
- Memory Aging and Cognition Centre, National University Health System
| |
Collapse
|
56
|
Machado V, Gilsbach R, Das R, Schober A, Bogatyreva L, Hauschke D, Krieglstein K, Unsicker K, Spittau B. Gdf-15 deficiency does not alter vulnerability of nigrostriatal dopaminergic system in MPTP-intoxicated mice. Cell Tissue Res 2016; 365:209-23. [PMID: 27115420 DOI: 10.1007/s00441-016-2406-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/06/2016] [Indexed: 12/25/2022]
Abstract
Growth/differentiation factor-15 (Gdf-15) is a member of the transforming growth factor-β (Tgf-β) superfamily and has been shown to be a potent neurotrophic factor for midbrain dopaminergic (DAergic) neurons both in vitro and in vivo. Gdf-15 has also been shown to be involved in inflammatory processes. The aim of this study was to identify the role of endogenous Gdf-15 in the MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) mouse model of Parkinson's disease (PD) by comparing Gdf-15 (+/+) and Gdf-15 (-/-) mice. At 4 days and 14 days post-MPTP administration, both Gdf-15 (+/+) and Gdf-15 (-/-) mice showed a similar decline in DAergic neuron numbers and in striatal dopamine (DA) levels. This was followed by a comparable restorative phase at 90 days and 120 days, indicating that the absence of Gdf-15 does not affect the susceptibility or the recovery capacity of the nigrostriatal system after MPTP administration. The MPTP-induced microglial and astrocytic response was not significantly altered between the two genotypes. However, pro-inflammatory and anti-inflammatory cytokine profiling revealed the differential expression of markers in Gdf-15 (+/+) and Gdf-15 (-/-) mice after MPTP administration. Thus, the MPTP mouse model fails to uncover a major role of endogenous Gdf-15 in the protection of MPTP-lesioned nigrostriatal DAergic neurons, in contrast to its capacity to protect the 6-hydroxydopamine-intoxicated nigrostriatal system.
Collapse
Affiliation(s)
- Venissa Machado
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104, Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, 79104, Freiburg, Germany
| | - Richa Das
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany.,German Center for Neurodegenerative Diseases, 53115, Bonn, Germany
| | - Andreas Schober
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany
| | - Lioudmila Bogatyreva
- Institute of Medical Biometry and Medical Informatics, University of Freiburg, 79104, Freiburg, Germany
| | - Dieter Hauschke
- German Center for Neurodegenerative Diseases, 53115, Bonn, Germany
| | - Kerstin Krieglstein
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany
| | - Klaus Unsicker
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany.
| | - Björn Spittau
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
57
|
GDF-15 enhances intracellular Ca2+ by increasing Cav1.3 expression in rat cerebellar granule neurons. Biochem J 2016; 473:1895-904. [PMID: 27114559 PMCID: PMC4925162 DOI: 10.1042/bcj20160362] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 04/25/2016] [Indexed: 12/17/2022]
Abstract
GDF-15 (growth/differentiation factor 15) is a novel member of the TGF (transforming growth factor)-β superfamily that has critical roles in the central and peripheral nervous systems. We reported previously that GDF-15 increased delayed rectifier outward K+ currents and Kv2.1 α subunit expression through TβRII (TGF-β receptor II) to activate Src kinase and Akt/mTOR (mammalian target of rapamycin) signalling in rat CGNs (cerebellar granule neurons). In the present study, we found that treatment of CGNs with GDF-15 for 24 h increased the intracellular Ca2+ concentration ([Ca2+]i) in response to membrane depolarization, as determined by Ca2+ imaging. Whole-cell current recordings indicated that GDF-15 increased the inward Ca2+ current (ICa) without altering steady-state activation of Ca2+ channels. Treatment with nifedipine, an inhibitor of L-type Ca2+ channels, abrogated GDF-15-induced increases in [Ca2+]i and ICa. The GDF-15-induced increase in ICa was mediated via up-regulation of the Cav1.3 α subunit, which was attenuated by inhibiting Akt/mTOR and ERK (extracellular-signal-regulated kinase) pathways and by pharmacological inhibition of Src-mediated TβRII phosphorylation. Given that Cav1.3 is not only a channel for Ca2+ influx, but also a transcriptional regulator, our data confirm that GDF-15 induces protein expression via TβRII and activation of a non-Smad pathway, and provide novel insight into the mechanism of GDF-15 function in neurons.
Collapse
|
58
|
Fujita Y, Taniguchi Y, Shinkai S, Tanaka M, Ito M. Secreted growth differentiation factor 15 as a potential biomarker for mitochondrial dysfunctions in aging and age-related disorders. Geriatr Gerontol Int 2016; 16 Suppl 1:17-29. [DOI: 10.1111/ggi.12724] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2015] [Indexed: 12/28/2022]
Affiliation(s)
- Yasunori Fujita
- Research Teams for; Mechanism of Aging; Tokyo Metropolitan Institute of Gerontology; Tokyo Japan
| | - Yu Taniguchi
- Social Participation and Community Health; Tokyo Metropolitan Institute of Gerontology; Tokyo Japan
| | - Shoji Shinkai
- Social Participation and Community Health; Tokyo Metropolitan Institute of Gerontology; Tokyo Japan
| | - Masashi Tanaka
- Department of Genomics for Longevity and Health; Tokyo Metropolitan Institute of Gerontology; Tokyo Japan
| | - Masafumi Ito
- Research Teams for; Mechanism of Aging; Tokyo Metropolitan Institute of Gerontology; Tokyo Japan
| |
Collapse
|
59
|
Li C, Wang X, Casal I, Wang J, Li P, Zhang W, Xu E, Lai M, Zhang H. Growth differentiation factor 15 is a promising diagnostic and prognostic biomarker in colorectal cancer. J Cell Mol Med 2016; 20:1420-6. [PMID: 26990020 PMCID: PMC4956952 DOI: 10.1111/jcmm.12830] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 02/05/2016] [Indexed: 12/21/2022] Open
Abstract
Although various studies have demonstrated that growth differentiation factor 15 (GDF15) might be a potential diagnostic and prognostic marker in colorectal cancer (CRC) patients, the results are inconsistent and the statistical power of individual studies is also insufficient. An original study was conducted to explore the diagnostic and prognostic value of serum GDF15 in CRC patients. We also conducted a meta‐analysis study which aimed to summarize the diagnostic and prognostic performance of serum GDF15 in CRC. We searched PubMed and ISI Web of Knowledge up to 1 November 2014 for eligible studies. In order to explore the diagnostic performance of GDF15, standardized mean difference (SMD) and their 95% confidence intervals (CI) were estimated and receiver‐operating characteristic (ROC) curves were constructed. For prognostic meta‐analysis, study‐specific hazard ratios (HRs) of serum GDF15 for survival were summarized. A total of eight studies were included in the meta‐analyses. Our results revealed that serum GDF15 levels in CRC patients were higher than those in healthy controls (SMD = 1.08, 95% CI: 0.56–1.59, P < 0.001). For discriminating CRC from healthy controls, the AUC of GDF15 was 0.816 (95% CI: 0.792–0.838). The sensitivity and specificity were 58.9% (95% CI: 55.0–62.8) and 92.08% (95% CI: 89.2–94.4), respectively, when a cut‐off value of 1099 pg/ml was established. Besides, higher GDF15 expression level was associated with worse overall survival for CRC patients (pooled HR = 2.09, 95% CI: 1.47–2.96). In conclusion, the present meta‐analysis suggests that serum GDF15 may be a useful diagnostic and prognostic biomarker for CRC.
Collapse
Affiliation(s)
- Chen Li
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, China
| | - Xiaobing Wang
- Medical Center for Tumor Detection, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ignacio Casal
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - Jingyu Wang
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Pathology, the First Hospital of Jiaxing, Jiaxing, China
| | - Peiwei Li
- Department of Epidemiology and Health Statistics, Zhejiang University School of Public Health, Hangzhou, China
| | - Wei Zhang
- Medical Center for Tumor Detection, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Enping Xu
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, China
| | - Maode Lai
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, China
| | - Honghe Zhang
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, China
| |
Collapse
|
60
|
Machado V, Haas SJP, von Bohlen Und Halbach O, Wree A, Krieglstein K, Unsicker K, Spittau B. Growth/differentiation factor-15 deficiency compromises dopaminergic neuron survival and microglial response in the 6-hydroxydopamine mouse model of Parkinson's disease. Neurobiol Dis 2015; 88:1-15. [PMID: 26733415 DOI: 10.1016/j.nbd.2015.12.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/21/2015] [Accepted: 12/25/2015] [Indexed: 12/15/2022] Open
Abstract
Growth/differentiation factor-15 (Gdf-15) is a member of the TGF-β superfamily and a pleiotropic, widely distributed cytokine, which has been shown to play roles in various pathologies, including inflammation. Analysis of Gdf-15(-/-) mice has revealed that it serves the postnatal maintenance of spinal cord motor neurons and sensory neurons. In a previous study, exogenous Gdf-15 rescued 6-hydroxydopamine (6-OHDA) lesioned Gdf-15(+/+) nigrostriatal dopaminergic (DAergic) neurons in vitro and in vivo. Whether endogenous Gdf-15 serves the physiological maintenance of nigrostriatal DAergic neurons in health and disease is not known and was addressed in the present study. Stereotactic injection of 6-OHDA into the medial forebrain bundle (MFB) led to a significant decline in the numbers of DAergic neurons in both Gdf-15(+/+) and Gdf-15(-/-) mice over a time-period of 14days. However, this decrease was exacerbated in the Gdf-15(-/-) mice, with only 5.5% surviving neurons as compared to 24% in the Gdf-15(+/+) mice. Furthermore, the microglial response to the 6-OHDA lesion was reduced in Gdf-15(-/-) mice, with significantly lower numbers of total and activated microglia and a differential cytokine expression as compared to the Gdf-15(+/+) mice. Using in vitro models, we could demonstrate the importance of endogenous Gdf-15 in promoting DAergic neuron survival thus highlighting its relevance in a direct neurotrophic supportive role. Taken together, these results indicate the importance of Gdf-15 in promoting survival of DAergic neurons and regulating the inflammatory response post 6-OHDA lesion.
Collapse
Affiliation(s)
- Venissa Machado
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.
| | - Stefan J-P Haas
- Department of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany.
| | | | - Andreas Wree
- Department of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany.
| | - Kerstin Krieglstein
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104 Freiburg, Germany.
| | - Klaus Unsicker
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104 Freiburg, Germany.
| | - Björn Spittau
- Institute of Anatomy and Cell Biology, Department of Molecular Embryology, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
61
|
Jiang J, Trollor JN, Brown DA, Crawford JD, Thalamuthu A, Smith E, Breit SN, Liu T, Brodaty H, Baune BT, Sachdev PS, Wen W. An inverse relationship between serum macrophage inhibitory cytokine-1 levels and brain white matter integrity in community-dwelling older individuals. Psychoneuroendocrinology 2015; 62:80-8. [PMID: 26254771 DOI: 10.1016/j.psyneuen.2015.07.610] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 07/17/2015] [Accepted: 07/23/2015] [Indexed: 11/28/2022]
Abstract
Macrophage inhibitory cytokine-1 (MIC-1/GDF15) is a marker of inflammation that has been associated with atherosclerosis. We have previously demonstrated its relationships with cognitive decline and cerebral gray matter volumes, suggesting its role as a biomarker of cognitive impairment. Considering that it is widely distributed in the brain, and both inflammation and vascular pathology impact on white matter (WM) integrity, we examined the relationship between MIC-1/GDF15 and measures of WM integrity, including WM volumes, mean fractional anisotropy (FA) values and WM hyperintensity (WMH) volumes in a community-dwelling non-demented sample of older individuals (n=327, 70-90 years old). We found that the mean FA values were negatively associated with MIC-1/GDF15 serum levels, after Bonferroni correction. The voxel-wise analysis showed negative relationships between MIC-1/GDF15 serum levels and FA values in corticospinal tract, corpus callosum (including genu, body and splenium parts), superior longitudinal fasciculus, cingulum, as well as anterior and posterior thalamic radiation. Whole brain WMH volumes, especially deep WMH volumes, showed a non-significant trend for a positive association with MIC-1/GDF15 serum levels. The associations between MIC-1/GDF15 serum levels and WM integrity showed a non-significant trend of being stronger for the individuals classified as mild cognitive impairment, compared to the normal ageing participants. The findings suggest that high serum MIC-1/GDF15 levels indicate reduced WM integrity and possibly greater WM pathology.
Collapse
Affiliation(s)
- Jiyang Jiang
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick, NSW, Australia
| | - Julian N Trollor
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick, NSW, Australia; Department of Development Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - David A Brown
- Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Darlinghurst, NSW, Australia
| | - John D Crawford
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick, NSW, Australia
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick, NSW, Australia
| | - Evelyn Smith
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick, NSW, Australia
| | - Samuel N Breit
- Centre for Applied Medical Research, St. Vincent's Hospital and University of New South Wales, Darlinghurst, NSW, Australia
| | - Tao Liu
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Henry Brodaty
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick, NSW, Australia; Aged Care Psychiatry, Prince of Wales Hospital, Randwick, NSW, Australia; Dementia Collaborative Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Bernhard T Baune
- Discipline of Psychiatry, School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick, NSW, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Wei Wen
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick, NSW, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Randwick, NSW, Australia.
| |
Collapse
|
62
|
Yadin D, Knaus P, Mueller TD. Structural insights into BMP receptors: Specificity, activation and inhibition. Cytokine Growth Factor Rev 2015; 27:13-34. [PMID: 26690041 DOI: 10.1016/j.cytogfr.2015.11.005] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 11/13/2015] [Indexed: 12/29/2022]
Abstract
Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-β family (TGFβ), which signal through hetero-tetrameric complexes of type I and type II receptors. In humans there are many more TGFβ ligands than receptors, leading to the question of how particular ligands can initiate specific signaling responses. Here we review structural features of the ligands and receptors that contribute to this specificity. Ligand activity is determined by receptor-ligand interactions, growth factor prodomains, extracellular modulator proteins, receptor assembly and phosphorylation of intracellular signaling proteins, including Smad transcription factors. Detailed knowledge about the receptors has enabled the development of BMP-specific type I receptor kinase inhibitors. In future these may help to treat human diseases such as fibrodysplasia ossificans progressiva.
Collapse
Affiliation(s)
- David Yadin
- Institute for Chemistry and Biochemistry, Free University Berlin, Institute of Chemistry and Biochemistry, D-14195 Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Campus Virchow Klinikum, Augustenburger Platz 1, D-13351 Berlin, Germany.
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Free University Berlin, Institute of Chemistry and Biochemistry, D-14195 Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Campus Virchow Klinikum, Augustenburger Platz 1, D-13351 Berlin, Germany.
| | - Thomas D Mueller
- Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute of the University Wuerzburg, Julius-von-Sachs-Platz 2, D-97082 Wuerzburg, Germany.
| |
Collapse
|
63
|
Kim DH, Lee D, Chang EH, Kim JH, Hwang JW, Kim JY, Kyung JW, Kim SH, Oh JS, Shim SM, Na DL, Oh W, Chang JW. GDF-15 secreted from human umbilical cord blood mesenchymal stem cells delivered through the cerebrospinal fluid promotes hippocampal neurogenesis and synaptic activity in an Alzheimer's disease model. Stem Cells Dev 2015; 24:2378-90. [PMID: 26154268 DOI: 10.1089/scd.2014.0487] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Our previous studies demonstrated that transplantation of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) into the hippocampus of a transgenic mouse model of Alzheimer's disease (AD) reduced amyloid-β (Aβ) plaques and enhanced cognitive function through paracrine action. Due to the limited life span of hUCB-MSCs after their transplantation, the extension of hUCB-MSC efficacy was essential for AD treatment. In this study, we show that repeated cisterna magna injections of hUCB-MSCs activated endogenous hippocampal neurogenesis and significantly reduced Aβ42 levels. To identify the paracrine factors released from the hUCB-MSCs that stimulated endogenous hippocampal neurogenesis in the dentate gyrus, we cocultured adult mouse neural stem cells (NSCs) with hUCB-MSCs and analyzed the cocultured media with cytokine arrays. Growth differentiation factor-15 (GDF-15) levels were significantly increased in the media. GDF-15 suppression in hUCB-MSCs with GDF-15 small interfering RNA reduced the proliferation of NSCs in cocultures. Conversely, recombinant GDF-15 treatment in both in vitro and in vivo enhanced hippocampal NSC proliferation and neuronal differentiation. Repeated administration of hUBC-MSCs markedly promoted the expression of synaptic vesicle markers, including synaptophysin, which are downregulated in patients with AD. In addition, in vitro synaptic activity through GDF-15 was promoted. Taken together, these results indicated that repeated cisterna magna administration of hUCB-MSCs enhanced endogenous adult hippocampal neurogenesis and synaptic activity through a paracrine factor of GDF-15, suggesting a possible role of hUCB-MSCs in future treatment strategies for AD.
Collapse
Affiliation(s)
- Dong Hyun Kim
- 1 Biomedical Research Institute , MEDIPOST Co., Ltd., Gyeonggi-Do, Republic of Korea.,2 Department of Biotechnology and Bioengineering, Sungkyunkwan University , Suwon, Republic of Korea
| | - Dahm Lee
- 1 Biomedical Research Institute , MEDIPOST Co., Ltd., Gyeonggi-Do, Republic of Korea
| | - Eun Hyuk Chang
- 3 Samsung Biomedical Research Institute, Samsung Advanced Institute of Technology , Samsung Electronics Co., Ltd., Seoul, Republic of Korea.,4 Department of Neurology, Samsung Medical Center, School of Medicine, Sungkyunkwan University , Seoul, Republic of Korea
| | - Ji Hyun Kim
- 1 Biomedical Research Institute , MEDIPOST Co., Ltd., Gyeonggi-Do, Republic of Korea.,4 Department of Neurology, Samsung Medical Center, School of Medicine, Sungkyunkwan University , Seoul, Republic of Korea
| | - Jung Won Hwang
- 4 Department of Neurology, Samsung Medical Center, School of Medicine, Sungkyunkwan University , Seoul, Republic of Korea.,5 Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University , Seoul, Republic of Korea
| | - Ju-Yeon Kim
- 1 Biomedical Research Institute , MEDIPOST Co., Ltd., Gyeonggi-Do, Republic of Korea
| | - Jae Won Kyung
- 6 Department of Neuroscience, Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University , Seoul, Republic of Korea
| | - Sung Hyun Kim
- 6 Department of Neuroscience, Neurodegeneration Control Research Center, School of Medicine, Kyung Hee University , Seoul, Republic of Korea
| | - Jeong Su Oh
- 2 Department of Biotechnology and Bioengineering, Sungkyunkwan University , Suwon, Republic of Korea
| | - Sang Mi Shim
- 7 Department of Biomedical Sciences, College of Medicine, Seoul National University , Seoul, Republic of Korea
| | - Duk Lyul Na
- 4 Department of Neurology, Samsung Medical Center, School of Medicine, Sungkyunkwan University , Seoul, Republic of Korea
| | - Wonil Oh
- 1 Biomedical Research Institute , MEDIPOST Co., Ltd., Gyeonggi-Do, Republic of Korea
| | - Jong Wook Chang
- 5 Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University , Seoul, Republic of Korea.,8 Stem Cell & Regenerative Medicine Center (SCRMC), Research Institute for Future Medicine , Samsung Medical Center, Seoul, Republic of Korea
| |
Collapse
|
64
|
Andersson C, Preis SR, Beiser A, DeCarli C, Wollert KC, Wang TJ, Januzzi JL, Vasan RS, Seshadri S. Associations of Circulating Growth Differentiation Factor-15 and ST2 Concentrations With Subclinical Vascular Brain Injury and Incident Stroke. Stroke 2015. [PMID: 26219649 DOI: 10.1161/strokeaha.115.009026] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Growth differentiation factor-15 (GDF-15) and soluble (s)ST2 are markers of cardiac and vascular stress. We investigated the associations between circulating concentrations of these biomarkers and incident stroke and subclinical vascular brain injury in a sample from the Framingham Offspring cohort. METHODS We followed 3374 stroke- and dementia-free individuals (mean age, 59.0±9.7 years; 53% women) attending the Framingham Offspring sixth examination cycle 11.8±3.0 years for incident stroke. A subsample of 2463 individuals underwent brain magnetic resonance imaging and neuropsychological testing ≈4.0±1.7 years after the sixth examination. RESULTS After adjustment for traditional cardiovascular risk factors, B-type natriuretic peptide, high-sensitivity C-reactive protein, and urine albumin levels, higher stress biomarker levels were associated cross-sectionally with lower brain volumes (β coefficients for intracranial volume comparing fourth [Q4] versus first biomarker [Q1] quartiles: -0.71% for GDF-15; P=0.002 and 0.47% for sST2; P=0.02) and worse performance on the visual reproduction test (β coefficients for Q4 versus Q1: -0.62 for GDF-15; P=0.009 and -0.40 for sST2; P=0.04). Higher GDF-15 concentrations were also associated with greater log-transformed white-matter hyperintensity volumes (β for Q4 versus Q1=0.19; P=0.01). Prospectively, a total of 203 (6%) individuals developed incident stroke/transient ischemic attack during follow-up. After multivariable adjustment, sST2 remained significantly associated with stroke/transient ischemic attack, hazard ratio for Q4 versus Q1 of 1.76, 95% confidence interval of 1.06 to 2.92, and P=0.03. CONCLUSIONS Circulating GDF-15 and sST2 are associated with subclinical brain injury and cognitive impairment. Higher sST2 concentrations are also associated with incident stroke, suggesting potential links between cardiac stress biomarkers and brain injury.
Collapse
Affiliation(s)
- Charlotte Andersson
- From the Framingham Heart Study, MA (C.A., A.B., T.J.W., R.S.V., S.S.); Department of Cardiology, Gentofte Hospital, University of Copenhagen, Denmark (C.A.); Department of Biostatistics, Boston University School of Public Health, MA (S.R.P., A.B.); Department of Neurology (A.B., S.S.) and Section of Preventive Medicine and Cardiology (R.S.V.), Boston University School of Medicine, MA; Department of Neurology, University of California at Davis, Sacramento (C.D.); Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany (K.C.W.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston (J.L.J.).
| | - Sarah R Preis
- From the Framingham Heart Study, MA (C.A., A.B., T.J.W., R.S.V., S.S.); Department of Cardiology, Gentofte Hospital, University of Copenhagen, Denmark (C.A.); Department of Biostatistics, Boston University School of Public Health, MA (S.R.P., A.B.); Department of Neurology (A.B., S.S.) and Section of Preventive Medicine and Cardiology (R.S.V.), Boston University School of Medicine, MA; Department of Neurology, University of California at Davis, Sacramento (C.D.); Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany (K.C.W.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston (J.L.J.)
| | - Alexa Beiser
- From the Framingham Heart Study, MA (C.A., A.B., T.J.W., R.S.V., S.S.); Department of Cardiology, Gentofte Hospital, University of Copenhagen, Denmark (C.A.); Department of Biostatistics, Boston University School of Public Health, MA (S.R.P., A.B.); Department of Neurology (A.B., S.S.) and Section of Preventive Medicine and Cardiology (R.S.V.), Boston University School of Medicine, MA; Department of Neurology, University of California at Davis, Sacramento (C.D.); Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany (K.C.W.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston (J.L.J.)
| | - Charles DeCarli
- From the Framingham Heart Study, MA (C.A., A.B., T.J.W., R.S.V., S.S.); Department of Cardiology, Gentofte Hospital, University of Copenhagen, Denmark (C.A.); Department of Biostatistics, Boston University School of Public Health, MA (S.R.P., A.B.); Department of Neurology (A.B., S.S.) and Section of Preventive Medicine and Cardiology (R.S.V.), Boston University School of Medicine, MA; Department of Neurology, University of California at Davis, Sacramento (C.D.); Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany (K.C.W.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston (J.L.J.)
| | - Kai C Wollert
- From the Framingham Heart Study, MA (C.A., A.B., T.J.W., R.S.V., S.S.); Department of Cardiology, Gentofte Hospital, University of Copenhagen, Denmark (C.A.); Department of Biostatistics, Boston University School of Public Health, MA (S.R.P., A.B.); Department of Neurology (A.B., S.S.) and Section of Preventive Medicine and Cardiology (R.S.V.), Boston University School of Medicine, MA; Department of Neurology, University of California at Davis, Sacramento (C.D.); Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany (K.C.W.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston (J.L.J.)
| | - Thomas J Wang
- From the Framingham Heart Study, MA (C.A., A.B., T.J.W., R.S.V., S.S.); Department of Cardiology, Gentofte Hospital, University of Copenhagen, Denmark (C.A.); Department of Biostatistics, Boston University School of Public Health, MA (S.R.P., A.B.); Department of Neurology (A.B., S.S.) and Section of Preventive Medicine and Cardiology (R.S.V.), Boston University School of Medicine, MA; Department of Neurology, University of California at Davis, Sacramento (C.D.); Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany (K.C.W.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston (J.L.J.)
| | - James L Januzzi
- From the Framingham Heart Study, MA (C.A., A.B., T.J.W., R.S.V., S.S.); Department of Cardiology, Gentofte Hospital, University of Copenhagen, Denmark (C.A.); Department of Biostatistics, Boston University School of Public Health, MA (S.R.P., A.B.); Department of Neurology (A.B., S.S.) and Section of Preventive Medicine and Cardiology (R.S.V.), Boston University School of Medicine, MA; Department of Neurology, University of California at Davis, Sacramento (C.D.); Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany (K.C.W.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston (J.L.J.)
| | - Ramachandran S Vasan
- From the Framingham Heart Study, MA (C.A., A.B., T.J.W., R.S.V., S.S.); Department of Cardiology, Gentofte Hospital, University of Copenhagen, Denmark (C.A.); Department of Biostatistics, Boston University School of Public Health, MA (S.R.P., A.B.); Department of Neurology (A.B., S.S.) and Section of Preventive Medicine and Cardiology (R.S.V.), Boston University School of Medicine, MA; Department of Neurology, University of California at Davis, Sacramento (C.D.); Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany (K.C.W.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston (J.L.J.)
| | - Sudha Seshadri
- From the Framingham Heart Study, MA (C.A., A.B., T.J.W., R.S.V., S.S.); Department of Cardiology, Gentofte Hospital, University of Copenhagen, Denmark (C.A.); Department of Biostatistics, Boston University School of Public Health, MA (S.R.P., A.B.); Department of Neurology (A.B., S.S.) and Section of Preventive Medicine and Cardiology (R.S.V.), Boston University School of Medicine, MA; Department of Neurology, University of California at Davis, Sacramento (C.D.); Division of Molecular and Translational Cardiology, Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany (K.C.W.); Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN (T.J.W.); and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston (J.L.J.)
| |
Collapse
|
65
|
Abstract
Bone morphogenetic proteins (BMPs), together with the eponymous transforming growth factor (TGF) β and the Activins form the TGFβ superfamily of ligands. This protein family comprises more than 30 structurally highly related proteins, which determine formation, maintenance, and regeneration of tissues and organs. Their importance for the development of multicellular organisms is evident from their existence in all vertebrates as well as nonvertebrate animals. From their highly specific functions in vivo either a strict relation between a particular ligand and its cognate cellular receptor and/or a stringent regulation to define a distinct temperospatial expression pattern for the various ligands and receptor is expected. However, only a limited number of receptors are found to serve a large number of ligands thus implicating highly promiscuous ligand-receptor interactions instead. Since in tissues a multitude of ligands are often found, which signal via a highly overlapping set of receptors, this raises the question how such promiscuous interactions between different ligands and their receptors can generate concerted and highly specific cellular signals required during embryonic development and tissue homeostasis.
Collapse
Affiliation(s)
- Thomas D Mueller
- Department Plant Physiology and Biophysics, Julius-von-Sachs Institute of the University Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|
66
|
Wang X, Krebbers J, Charalambous P, Machado V, Schober A, Bosse F, Müller HW, Unsicker K. Growth/differentiation factor-15 and its role in peripheral nervous system lesion and regeneration. Cell Tissue Res 2015; 362:317-30. [DOI: 10.1007/s00441-015-2219-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/20/2015] [Indexed: 01/31/2023]
|
67
|
GDF15 derived from both tumor-associated macrophages and esophageal squamous cell carcinomas contributes to tumor progression via Akt and Erk pathways. J Transl Med 2015; 95:491-503. [PMID: 25730371 DOI: 10.1038/labinvest.2015.36] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 12/09/2014] [Accepted: 12/09/2014] [Indexed: 12/21/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are known to be involved in the progression, angiogenesis, and motility of various cancers. We previously reported the association between an increased number of infiltrating TAMs with tumor progression and poor prognosis in esophageal squamous cell carcinomas (ESCCs). To study the roles of TAMs in ESCC, we first exposed peripheral blood monocyte (PBMo)-derived macrophages from healthy volunteers to conditioned media of TE series human ESCC cell line (TECM) and confirmed the induction of the expression of the M2 macrophage marker CD204 and the protumorigenic factors interleukin (IL)-10, VEGFA, and MMPs. Next, we compared gene expression profiles between PBMo-derived macrophages stimulated with or without TECM by cDNA microarray and focused on growth differentiation factor 15 (GDF15) among the highly expressed genes including IL-6, IL-8, and CXCL1. Our immunohistochemical study of 70 surgically resected ESCCs revealed that GDF15 was present not only in cancer cells but also in macrophages. The high expression of GDF15 in the ESCCs was significantly correlated with several more malignant phenotypes including vessel invasion, lymph node metastasis, and clinical stages. Patients with high GDF15 expression showed significantly poorer disease-free survival (P=0.011) and overall survival (P=0.041). We also found that recombinant human GDF15 promotes cell proliferation and the phosphorylation of both Akt and Erk1/2 in ESCC cell lines in vitro. These results indicate that GDF15 is secreted by both TAMs and cancer cells in the tumor microenvironment and is associated with aberrant growth and a poor prognosis in human ESCC.
Collapse
|
68
|
Jiang J, Wen W, Brown DA, Crawford J, Thalamuthu A, Smith E, Breit SN, Liu T, Zhu W, Brodaty H, Baune BT, Trollor JN, Sachdev PS. The relationship of serum macrophage inhibitory cytokine-1 levels with gray matter volumes in community-dwelling older individuals. PLoS One 2015; 10:e0123399. [PMID: 25867953 PMCID: PMC4395016 DOI: 10.1371/journal.pone.0123399] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 02/18/2015] [Indexed: 12/29/2022] Open
Abstract
Using circulating inflammatory markers and magnetic resonance imaging (MRI), recent studies have associated inflammation with brain volumetric measures. Macrophage Inhibitory Cytokine-1 (MIC-1/GDF15) is a divergent transforming growth factor - beta (TGF-β) superfamily cytokine. To uncover the underlying mechanisms of the previous finding of a negative association between MIC-1/GDF15 serum levels and cognition, the present study aimed to examine the relationship of circulating MIC-1/GDF15 levels with human brain gray matter (GM) volumes, in a community-dwelling sample aged 70-90 years over two years (Wave 1: n = 506, Wave 2: n = 327), of which the age-related brain atrophy had been previously well defined. T1-weighted MRI scans were obtained at both waves and analyzed using the FMRIB Software Library and FreeSurfer. The results showed significantly negative associations between MIC-1/GDF15 serum levels and both subcortical and cortical GM volumes. GM volumes of the whole brain, cortex, temporal lobe, thalamus and accumbens showed significant mediating effects on the associations between MIC-1/GDF15 serum levels and global cognition scores. Increases in MIC-1/GDF15 serum levels were associated with decreases in cortical and subcortical GM volume over two years. In conclusion, MIC-1/GDF15 serum levels were inversely associated with GM volumes both cross-sectionally and longitudinally.
Collapse
Affiliation(s)
- Jiyang Jiang
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick NSW, Australia
| | - Wei Wen
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick NSW, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Randwick NSW, Australia
- * E-mail:
| | - David A. Brown
- Centre for Applied Medical Research, St. Vincent’s Hospital and University of New South Wales, Darlinghurst NSW, Australia
| | - John Crawford
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick NSW, Australia
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick NSW, Australia
| | - Evelyn Smith
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick NSW, Australia
| | - Samuel N. Breit
- Centre for Applied Medical Research, St. Vincent’s Hospital and University of New South Wales, Darlinghurst NSW, Australia
| | - Tao Liu
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Wanlin Zhu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Henry Brodaty
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick NSW, Australia
- Aged Care Psychiatry, Prince of Wales Hospital, Randwick NSW, Australia
- Dementia Collaborative Research Centre, University of New South Wales, Sydney NSW, Australia
| | - Bernhard T. Baune
- Discipline of Psychiatry, School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Julian N. Trollor
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick NSW, Australia
- Department of Development Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney NSW, Australia
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, University of New South Wales, Randwick NSW, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Randwick NSW, Australia
| |
Collapse
|
69
|
Yi MH, Zhang E, Baek H, Kim S, Shin N, Kang JW, Lee S, Oh SH, Kim DW. Growth Differentiation Factor 15 Expression in Astrocytes After Excitotoxic Lesion in the Mouse Hippocampus. Exp Neurobiol 2015; 24:133-8. [PMID: 26113792 PMCID: PMC4479809 DOI: 10.5607/en.2015.24.2.133] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 02/16/2015] [Accepted: 02/21/2015] [Indexed: 12/16/2022] Open
Abstract
Growth differentiation factor 15 (GDF15) is, a member of the transforming growth factor β (TGF-β) superfamily of proteins. Although GDF15 is well established as a potent neurotrophic factor for neurons, little is known about its role in glial cells under neuropathological conditions. We monitored GDF15 expression in astrocyte activation after a kainic acid (KA)-induced neurodegeneration in the ICR mice hippocampus. In control, GDF15 immunoreactivity (IR) was evident in the neuronal layer of the hippocampus; however, GDF15 expression had increased in activated astrocytes throughout the hippocampal region at day 3 after the treatment with KA. LPS treatment in astrocytes dramatically increased GDF15 expression in primary astrocytes. In addition, LPS treatment resulted in the decrease of the IκB-α degradation and increase of the phosphorylation level of RelA/p65. These results indicate that GDF15 has a potential link to NF-κB activation, making GDF15 a valuable target for modulating inflammatory conditions.
Collapse
Affiliation(s)
- Min-Hee Yi
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 301-747, Korea
| | - Enji Zhang
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 301-747, Korea
| | - Hyunjung Baek
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 301-747, Korea. ; Department of Pediatrics, Chungnam National University Hospital, Daejeon 301-721, Korea
| | - Sena Kim
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 301-747, Korea. ; Department of Plastic Surgery, Chungnam National University Hospital, Daejeon 301-721, Korea
| | - Nara Shin
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 301-747, Korea. ; Department of Anesthesia and Pain Medicine, Chungnam National University Hospital, Daejeon 301-721, Korea
| | - Joon Won Kang
- Department of Pediatrics, Chungnam National University Hospital, Daejeon 301-721, Korea
| | - Sunyeul Lee
- Department of Anesthesia and Pain Medicine, Chungnam National University Hospital, Daejeon 301-721, Korea
| | - Sang-Ha Oh
- Department of Plastic Surgery, Chungnam National University Hospital, Daejeon 301-721, Korea
| | - Dong Woon Kim
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon 301-747, Korea
| |
Collapse
|
70
|
Adela R, Banerjee SK. GDF-15 as a Target and Biomarker for Diabetes and Cardiovascular Diseases: A Translational Prospective. J Diabetes Res 2015; 2015:490842. [PMID: 26273671 PMCID: PMC4530250 DOI: 10.1155/2015/490842] [Citation(s) in RCA: 331] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 01/19/2015] [Accepted: 01/20/2015] [Indexed: 12/20/2022] Open
Abstract
Growth differentiation factor-15 (GDF-15) is a stress responsive cytokine. It is highly expressed in cardiomyocytes, adipocytes, macrophages, endothelial cells, and vascular smooth muscle cells in normal and pathological condition. GDF-15 increases during tissue injury and inflammatory states and is associated with cardiometabolic risk. Increased GDF-15 levels are associated with cardiovascular diseases such as hypertrophy, heart failure, atherosclerosis, endothelial dysfunction, obesity, insulin resistance, diabetes, and chronic kidney diseases in diabetes. Increased GDF-15 level is linked with the progression and prognosis of the disease condition. Age, smoking, and environmental factors are other risk factors that may increase GDF-15 level. Most of the scientific studies reported that GDF-15 plays a protective role in different tissues. However, few reports show that the deficiency of GDF-15 is beneficial against vascular injury and inflammation. GDF-15 protects heart, adipose tissue, and endothelial cells by inhibiting JNK (c-Jun N-terminal kinase), Bad (Bcl-2-associated death promoter), and EGFR (epidermal growth factor receptor) and activating Smad, eNOS, PI3K, and AKT signaling pathways. The present review describes the different animal and clinical studies and patent updates of GDF-15 in diabetes and cardiovascular diseases. It is a challenge for the scientific community to use GDF-15 information for patient monitoring, clinical decision-making, and replacement of current treatment strategies for diabetic and cardiovascular diseases.
Collapse
Affiliation(s)
- Ramu Adela
- Drug Discovery Research Center, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana 122014, India
| | - Sanjay K. Banerjee
- Drug Discovery Research Center, Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana 122014, India
- *Sanjay K. Banerjee:
| |
Collapse
|
71
|
GDF15 regulates Kv2.1-mediated outward K+ current through the Akt/mTOR signalling pathway in rat cerebellar granule cells. Biochem J 2014; 460:35-47. [PMID: 24597762 PMCID: PMC4000135 DOI: 10.1042/bj20140155] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
GDF15 (growth/differentiation factor 15), a novel member of the TGFβ (transforming growth factor β) superfamily, plays critical roles in the central and peripheral nervous systems, but the signal transduction pathways and receptor subtypes involved are not well understood. In the present paper, we report that GDF15 specifically increases the IK (delayed-rectifier outward K+ current) in rat CGNs (cerebellar granule neurons) in time- and concentration-dependent manners. The GDF15-induced amplification of the IK is mediated by the increased expression and reduced lysosome-dependent degradation of the Kv2.1 protein, the main α-subunit of the IK channel. Exposure of CGNs to GDF15 markedly induced the phosphorylation of ERK (extracellular-signal-regulated kinase), Akt and mTOR (mammalian target of rapamycin), but the GDF15-induced IK densities and increased expression of Kv2.1 were attenuated only by Akt and mTOR, and not ERK, inhibitors. Pharmacological inhibition of the Src-mediated phosphorylation of TGFβR2 (TGFβ receptor 2), not TGFβR1, abrogated the effect of GDF15 on IK amplification and Kv2.1 induction. Immunoprecipitation assays showed that GDF15 increased the tyrosine phosphorylation of TGFβRII in the CGN lysate. The results of the present study reveal a novel regulation of Kv2.1 by GDF15 mediated through the TGFβRII-activated Akt/mTOR pathway, which is a previously uncharacterized Smad-independent mechanism of GDF15 signalling.
Collapse
|
72
|
Carrillo-García C, Prochnow S, Simeonova IK, Strelau J, Hölzl-Wenig G, Mandl C, Unsicker K, von Bohlen Und Halbach O, Ciccolini F. Growth/differentiation factor 15 promotes EGFR signalling, and regulates proliferation and migration in the hippocampus of neonatal and young adult mice. Development 2014; 141:773-83. [PMID: 24496615 PMCID: PMC3930467 DOI: 10.1242/dev.096131] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The activation of epidermal growth factor receptor (EGFR) affects multiple aspects of neural precursor behaviour, including proliferation and migration. Telencephalic precursors acquire EGF responsiveness and upregulate EGFR expression at late stages of development. The events regulating this process and its significance are still unclear. We here show that in the developing and postnatal hippocampus (HP), growth/differentiation factor (GDF) 15 and EGFR are co-expressed in primitive precursors as well as in more differentiated cells. We also provide evidence that GDF15 promotes responsiveness to EGF and EGFR expression in hippocampal precursors through a mechanism that requires active CXC chemokine receptor (CXCR) 4. Besides EGFR expression, GDF15 ablation also leads to decreased proliferation and migration. In particular, lack of GDF15 impairs both processes in the cornu ammonis (CA) 1 and only proliferation in the dentate gyrus (DG). Importantly, migration and proliferation in the mutant HP were altered only perinatally, when EGFR expression was also affected. These data suggest that GDF15 regulates migration and proliferation by promoting EGFR signalling in the perinatal HP and represent a first description of a functional role for GDF15 in the developing telencephalon.
Collapse
Affiliation(s)
- Carmen Carrillo-García
- Department of Neurobiology, Interdisciplinary Centre for Neuroscience (IZN), Ruprecht-Karls University of Heidelberg, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Roles for the TGFβ superfamily in the development and survival of midbrain dopaminergic neurons. Mol Neurobiol 2014; 50:559-73. [PMID: 24504901 DOI: 10.1007/s12035-014-8639-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/02/2014] [Indexed: 12/29/2022]
Abstract
The adult midbrain contains 75% of all dopaminergic neurons in the CNS. Within the midbrain, these neurons are divided into three anatomically and functionally distinct clusters termed A8, A9 and A10. The A9 group plays a functionally non-redundant role in the control of voluntary movement, which is highlighted by the motor syndrome that results from their progressive degeneration in the neurodegenerative disorder, Parkinson's disease. Despite 50 years of investigation, treatment for Parkinson's disease remains symptomatic, but an intensive research effort has proposed delivering neurotrophic factors to the brain to protect the remaining dopaminergic neurons, or using these neurotrophic factors to differentiate dopaminergic neurons from stem cell sources for cell transplantation. Most neurotrophic factors studied in this context have been members of the transforming growth factor β (TGFβ) superfamily. In recent years, an intensive research effort has focused on understanding the function of these proteins in midbrain dopaminergic neuron development and their role in the molecular architecture that regulates the development of this brain region, with the goal of applying this knowledge to develop novel therapies for Parkinson's disease. In this review, the current evidence showing that TGFβ superfamily members play critical roles in the regulation of midbrain dopaminergic neuron induction, differentiation, target innervation and survival during embryonic and postnatal development is analysed, and the implications of these findings are discussed.
Collapse
|
74
|
Harvey A, Yen TY, Aizman I, Tate C, Case C. Proteomic analysis of the extracellular matrix produced by mesenchymal stromal cells: implications for cell therapy mechanism. PLoS One 2013; 8:e79283. [PMID: 24244468 PMCID: PMC3828366 DOI: 10.1371/journal.pone.0079283] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 09/27/2013] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) transiently transfected with notch1 intracellular domain (NICD) are beneficial for neurological disorders as observed in several preclinical studies. Extracellular matrix (ECM) derived from NICD-transfected MSCs has been previously shown to support in vitro neural cell growth and survival better than that of un-transfected MSCs. To understand the underlying mechanism(s) by which NICD-transfected MSC-derived ECM supports neural cell growth and survival, we investigated the differences in NICD-transfected MSC- and MSC-derived ECM protein quantity and composition. To compare the ECM derived from MSCs and NICD-transfected MSCs, the proteins were sequentially solubilized using sodium dodecyl sulfate (SDS) and urea, quantified, and compared across four human donors. We then analyzed ECM proteins using either in-gel digests or in-solution surfactant-assisted trypsin digests (SAISD) coupled with reverse phase nano-liquid chromatography and tandem mass spectrometry (nLC-MS/MS). Analyses using nLC-MS/MS identified key components of ECM from NICD-transfected MSCs and un-transfected MSCs and revealed significant differences in their respective compositions. This work provides a reproducible method for identifying and comparing in vitro cell-derived ECM proteins, which is crucial for exploring the mechanisms underlying cellular therapy.
Collapse
Affiliation(s)
- Adam Harvey
- SanBio Inc., Mountain View, California, United States of America
- Department of Biology, San Francisco State University, San Francisco, California, United States of America
| | - Ten-Yang Yen
- Department of Chemistry and Biochemistry, San Francisco State University, San Francisco, California, United States of America
| | - Irina Aizman
- SanBio Inc., Mountain View, California, United States of America
| | - Ciara Tate
- SanBio Inc., Mountain View, California, United States of America
| | - Casey Case
- SanBio Inc., Mountain View, California, United States of America
- * E-mail:
| |
Collapse
|
75
|
The multiple facets of the TGF-β family cytokine growth/differentiation factor-15/macrophage inhibitory cytokine-1. Cytokine Growth Factor Rev 2013; 24:373-84. [DOI: 10.1016/j.cytogfr.2013.05.003] [Citation(s) in RCA: 220] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 05/21/2013] [Indexed: 12/23/2022]
|
76
|
Charalambous P, Wang X, Thanos S, Schober A, Unsicker K. Regulation and effects of GDF-15 in the retina following optic nerve crush. Cell Tissue Res 2013; 353:1-8. [DOI: 10.1007/s00441-013-1634-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 04/10/2013] [Indexed: 12/21/2022]
|
77
|
Local substitution of GDF-15 improves axonal and sensory recovery after peripheral nerve injury. Cell Tissue Res 2012; 350:225-38. [DOI: 10.1007/s00441-012-1493-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 08/17/2012] [Indexed: 10/27/2022]
|
78
|
Promiscuity and specificity in BMP receptor activation. FEBS Lett 2012; 586:1846-59. [PMID: 22710174 DOI: 10.1016/j.febslet.2012.02.043] [Citation(s) in RCA: 232] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 02/28/2012] [Accepted: 02/29/2012] [Indexed: 02/03/2023]
Abstract
Bone Morphogenetic Proteins (BMPs), together with Transforming Growth Factor (TGF)-β and Activins/Inhibins constitute the TGF-β superfamily of ligands. This superfamily is formed by more than 30 structurally related secreted proteins. Since TGF-β members act as morphogens, either a strict relation between a particular ligand to a distinct cellular receptor and/or temporospatial expression patterns of ligands and receptors is expected. Instead, only a limited number of receptors exist implicating promiscuous interactions of ligands and receptors. Furthermore, in complex tissues a multitude of different ligands can be found, which signal via overlapping subsets of receptors. This raises the intriguing question how concerted interactions of different ligands and receptors generate highly specific cellular signals, which are required during development and tissue homeostasis.
Collapse
|
79
|
Ruschke K, Hiepen C, Becker J, Knaus P. BMPs are mediators in tissue crosstalk of the regenerating musculoskeletal system. Cell Tissue Res 2012; 347:521-44. [PMID: 22327483 DOI: 10.1007/s00441-011-1283-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 11/10/2011] [Indexed: 12/22/2022]
Abstract
The musculoskeletal system is a tight network of many tissues. Coordinated interplay at a biochemical level between tissues is essential for development and repair. Traumatic injury usually affects several tissues and represents a large challenge in clinical settings. The current demand for potent growth factors in such applications thus accompanies the keen interest in molecular mechanisms and orchestration of tissue formation. Of special interest are multitasking growth factors that act as signals in a variety of cell types, both in a paracrine and in an autocrine manner, thereby inducing cell differentiation and coordinating not only tissue assembly at specific sites but also maturation and homeostasis. We concentrate here on bone morphogenetic proteins (BMPs), which are important crosstalk mediators known for their irreplaceable roles in vertebrate development. The molecular crosstalk during embryonic musculoskeletal tissue formation is recapitulated in adult repair. BMPs act at different levels from the initiation to maturation of newly formed tissue. Interestingly, this is influenced by the spatiotemporal expression of different BMPs, their receptors and co-factors at the site of repair. Thus, the regenerative potential of BMPs needs to be evaluated in the context of highly connected tissues such as muscle and bone and might indeed be different in more poorly connected tissues such as cartilage. This highlights the need for an understanding of BMP signaling across tissues in order to eventually improve BMP regenerative potential in clinical applications. In this review, the distinct members of the BMP family and their individual contribution to musculoskeletal tissue repair are summarized by focusing on their paracrine and autocrine functions.
Collapse
Affiliation(s)
- Karen Ruschke
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | | | | |
Collapse
|
80
|
Kadowaki M, Yoshioka H, Kamitani H, Watanabe T, Wade PA, Eling TE. DNA methylation-mediated silencing of nonsteroidal anti-inflammatory drug-activated gene (NAG-1/GDF15) in glioma cell lines. Int J Cancer 2012; 130:267-77. [PMID: 21437897 PMCID: PMC3133861 DOI: 10.1002/ijc.26082] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 02/17/2011] [Indexed: 12/31/2022]
Abstract
Nonsteroidal anti-inflammatory drug-activated gene, NAG-1, a transforming growth factor-β member, is involved in tumor progression and development. The association between NAG-1 expression and development and progression of glioma has not been well defined. Glioblastoma cell lines have lower basal expression of NAG-1 than other gliomas and normal astrocytes. Most primary human gliomas have very low levels of NAG-1 expression. NAG-1 basal expression appeared to inversely correlate with tumor grade in glioma. Aberrant promoter hypermethylation is a common mechanism for silencing of tumor suppressor genes in cancer cells. In glioblastoma cell lines, NAG-1 expression was increased by the demethylating agent, 5-aza-2'-deoxycytidine. To investigate whether the NAG-1 gene was silenced by hypermethylation in glioblastoma, we examined DNA methylation status using genomic bisulfite sequencing. The NAG-1 promoter was densely methylated in several glioblastoma cell lines as well as in primary oligodendroglioma tumor samples, which have low basal expression of NAG-1. DNA methylation at two specific sites (-53 and +55 CpG sites) in the NAG-1 promoter was strongly associated with low NAG-1 expression. The methylation of the NAG-1 promoter at the -53 site blocks Egr-1 binding and thereby suppresses Nag-1 induction. Treatment of cells with low basal NAG-1 expression with NAG-1 inducer also did not increase NAG-1. Incubation with a demethylation chemical increased Nag-1 basal expression and subsequent incubation with a NAG-1 inducer increased NAG-1 expression. We concluded from these data that methylation of specific promoter sequences causes transcriptional silencing of the NAG-1 locus in glioma and may ultimately contribute to tumor progression.
Collapse
Affiliation(s)
- Mitsutoshi Kadowaki
- Laboratory of Molecular Carcinogenesis, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
- Division of Neurosurgery, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8504, Japan
| | - Hiroki Yoshioka
- Division of Neurosurgery, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8504, Japan
| | - Hideki Kamitani
- Division of Neurosurgery, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8504, Japan
| | - Takashi Watanabe
- Division of Neurosurgery, Institute of Neurological Sciences, Faculty of Medicine, Tottori University, Yonago, Tottori 683-8504, Japan
| | - Paul A. Wade
- Laboratory of Molecular Carcinogenesis, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Thomas E. Eling
- Laboratory of Molecular Carcinogenesis, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709
| |
Collapse
|
81
|
Worthmann H, Kempf T, Widera C, Tryc AB, Goldbecker A, Ma YT, Deb M, Tountopoulou A, Lambrecht J, Heeren M, Lichtinghagen R, Wollert KC, Weissenborn K. Growth Differentiation Factor 15 Plasma Levels and Outcome after Ischemic Stroke. Cerebrovasc Dis 2011; 32:72-8. [DOI: 10.1159/000328233] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 03/28/2011] [Indexed: 11/19/2022] Open
|
82
|
Schindowski K, von Bohlen und Halbach O, Strelau J, Ridder DA, Herrmann O, Schober A, Schwaninger M, Unsicker K. Regulation of GDF-15, a distant TGF-β superfamily member, in a mouse model of cerebral ischemia. Cell Tissue Res 2010; 343:399-409. [PMID: 21128084 PMCID: PMC3032194 DOI: 10.1007/s00441-010-1090-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 11/04/2010] [Indexed: 11/20/2022]
Abstract
GDF-15 is a novel distant member of the TGF-β superfamily and is widely distributed in the brain and peripheral nervous system. We have previously reported that GDF-15 is a potent neurotrophic factor for lesioned dopaminergic neurons in the substantia nigra, and that GDF-15-deficient mice show progressive postnatal losses of motor and sensory neurons. We have now investigated the regulation of GDF-15 mRNA and immunoreactivity in the murine hippocampal formation and selected cortical areas following an ischemic lesion by occlusion of the middle cerebral artery (MCAO). MCAO prominently upregulates GDF-15 mRNA in the hippocampus and parietal cortex at 3 h and 24 h after lesion. GDF-15 immunoreactivity, which is hardly detectable in the unlesioned brain, is drastically upregulated in neurons identified by double-staining with NeuN. NeuN staining reveals that most, if not all, neurons in the granular layer of the dentate gyrus and pyramidal layers of the cornu ammonis become GDF-15-immunoreactive. Moderate induction of GDF-15 immunoreactivity has been observed in a small number of microglial cells identified by labeling with tomato lectin, whereas astroglial cells remain GDF-15-negative after MCAO. Comparative analysis of the size of the infarcted area after MCAO in GDF-15 wild-type and knockout mice has failed to reveal significant differences. Together, our data substantiate the notion that GDF-15 is prominently upregulated in the lesioned brain and might be involved in orchestrating post-lesional responses other than the trophic support of neurons.
Collapse
Affiliation(s)
- Katharina Schindowski
- Institute for Pharmaceutical Biotechnology, University of Applied Science Biberach, Biberach/Riss, Germany
| | | | | | | | | | | | | | | |
Collapse
|
83
|
MIMEAULT MURIELLE, BATRA SURINDERK. Divergent molecular mechanisms underlying the pleiotropic functions of macrophage inhibitory cytokine-1 in cancer. J Cell Physiol 2010; 224:626-35. [PMID: 20578239 PMCID: PMC2932466 DOI: 10.1002/jcp.22196] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Multifunctional macrophage inhibitory cytokine-1, MIC-1, is a member of the transforming growth factor-beta (TGF-beta) superfamily that plays key roles in the prenatal development and regulation of the cellular responses to stress signals and inflammation and tissue repair after acute injuries in adult life. The stringent control of the MIC-1 expression, secretion, and functions involves complex regulatory mechanisms and the interplay of other growth factor signaling networks that control the cell behavior. The deregulation of MIC-1 expression and signaling pathways has been associated with diverse human diseases and cancer progression. The MIC-1 expression levels substantially increase in cancer cells, serum, and/or cerebrospinal fluid during the progression of diverse human aggressive cancers, such as intracranial brain tumors, melanoma, and lung, gastrointestinal, pancreatic, colorectal, prostate, and breast epithelial cancers. Of clinical interest, an enhanced MIC-1 expression has been positively correlated with poor prognosis and patient survival. Secreted MIC-1 cytokine, like the TGF-beta prototypic member of the superfamily, may provide pleiotropic roles in the early and late stages of carcinogenesis. In particular, MIC-1 may contribute to the proliferation, migration, invasion, metastases, and treatment resistance of cancer cells as well as tumor-induced anorexia and weight loss in the late stages of cancer. Thus, secreted MIC-1 cytokine constitutes a new potential biomarker and therapeutic target of great clinical interest for the development of novel diagnostic and prognostic methods and/or cancer treatment against numerous metastatic, recurrent, and lethal cancers.
Collapse
Affiliation(s)
- MURIELLE MIMEAULT
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska
| | - SURINDER K. BATRA
- Department of Biochemistry and Molecular Biology, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
84
|
Shnaper S, Desbaillets I, Brown DA, Murat A, Migliavacca E, Schluep M, Ostermann S, Hamou MF, Stupp R, Breit SN, de Tribolet N, Hegi ME. Elevated levels of MIC-1/GDF15 in the cerebrospinal fluid of patients are associated with glioblastoma and worse outcome. Int J Cancer 2009; 125:2624-30. [PMID: 19521960 DOI: 10.1002/ijc.24639] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
For patients with brain tumors identification of diagnostic and prognostic markers in easy accessible biological material, such as plasma or cerebrospinal fluid (CSF), would greatly facilitate patient management. MIC-1/GDF15 (growth differentiation factor 15) is a secreted protein of the TGF-beta superfamily and emerged as a candidate marker exhibiting increasing mRNA expression during malignant progression of glioma. Determination of MIC-1/GDF15 protein levels by ELISA in the CSF of a cohort of 94 patients with intracranial tumors including gliomas, meningioma and metastasis revealed significantly increased concentrations in glioblastoma patients (median, 229 pg/ml) when compared with control cohort of patients treated for non-neoplastic diseases (median below limit of detection of 156 pg/ml, p < 0.0001, Mann-Whitney test). However, plasma MIC-1/GDF15 levels were not elevated in the matching plasma samples from these patients. Most interestingly, patients with glioblastoma and increased CSF MIC-1/GDF15 had a shorter survival (p = 0.007, log-rank test). In conclusion, MIC-1/GDF15 protein measured in the CSF may have diagnostic and prognostic value in patients with intracranial tumors.
Collapse
Affiliation(s)
- Sophie Shnaper
- Department of Neurosurgery, Laboratory of Brain Tumor Biology and Genetics, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Alavian KN, Sgadò P, Alberi L, Subramaniam S, Simon HH. Elevated P75NTR expression causes death of engrailed-deficient midbrain dopaminergic neurons by Erk1/2 suppression. Neural Dev 2009; 4:11. [PMID: 19291307 PMCID: PMC2667502 DOI: 10.1186/1749-8104-4-11] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2008] [Accepted: 03/16/2009] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The homeodomain transcription factors Engrailed-1 and Engrailed-2 are required for the survival of mesencephalic dopaminergic (mesDA) neurons in a cell-autonomous and gene-dose-dependent manner. Homozygote mutant mice, deficient of both genes (En1-/-;En2-/-), die at birth and exhibit a loss of all mesDA neurons by mid-gestation. In heterozygote animals (En1+/-;En2-/-), which are viable and fertile, postnatal maintenance of the nigrostriatal dopaminergic system is afflicted, leading to a progressive degeneration specific to this subpopulation and Parkinson's disease-like molecular and behavioral deficits. RESULTS In this work, we show that the dose of Engrailed is inversely correlated to the expression level of the pan-neurotrophin receptor gene P75NTR (Ngfr). Loss of mesDA neurons in the Engrailed-null mutant embryos is caused by elevated expression of this neurotrophin receptor: Unusually, in this case, the cell death signal of P75NTR is mediated by suppression of Erk1/2 (extracellular-signal-regulated kinase 1/2) activity. The reduction in expression of Engrailed, possibly related to the higher levels of P75NTR, also decreases mitochondrial stability. In particular, the dose of Engrailed determines the sensitivity to cell death induced by the classic Parkinson-model toxin MPTP and to inhibition of the anti-apoptotic members of the Bcl-2 family of proteins. CONCLUSION Our study links the survival function of the Engrailed genes in developing mesDA neurons to the regulation of P75NTR and the sensitivity of these neurons to mitochondrial insult. The similarities to the disease etiology in combination with the nigral phenotype of En1+/-;En2-/- mice suggests that haplotype variations in the Engrailed genes and/or P75NTR that alter their expression levels could, in part, determine susceptibility to Parkinson's disease.
Collapse
Affiliation(s)
- Kambiz N Alavian
- Interdisciplinary Centre for Neuroscience, Department of Neuroanatomy, Ruprecht-Karls-Universität, 69120 Heidelberg, Germany
- Harvard Medical School, Neuroregeneration Labs, MRC 1, McLean Hospital, Mill St, Belmont, MA 02478, USA
| | - Paola Sgadò
- Interdisciplinary Centre for Neuroscience, Department of Neuroanatomy, Ruprecht-Karls-Universität, 69120 Heidelberg, Germany
- Paola Sgadò, Neurogenetics Laboratory, Child Neurology Unit, Pediatric Hospital A Meyer, Piazza di Careggi, 50139 Florence, Italy
| | - Lavinia Alberi
- Interdisciplinary Centre for Neuroscience, Department of Neuroanatomy, Ruprecht-Karls-Universität, 69120 Heidelberg, Germany
- The Johns Hopkins Institute for Cell Engineering, Department of Neurology, North Broadway Street, BRB 720, Baltimore, MD 2120, USA
| | - Srinivasa Subramaniam
- Interdisciplinary Centre for Neuroscience, Department of Neuroanatomy, Ruprecht-Karls-Universität, 69120 Heidelberg, Germany
- Department of Neuroscience, Johns Hopkins Medical School, N Wolfe Street, Baltimore, MD 21210, USA
| | - Horst H Simon
- Interdisciplinary Centre for Neuroscience, Department of Neuroanatomy, Ruprecht-Karls-Universität, 69120 Heidelberg, Germany
| |
Collapse
|
86
|
Strelau J, Schmeer C, Peterziel H, Sackmann T, Herold-Mende C, Steiner H, Weller M, Unsicker K. Expression and putative functions of GDF-15, a member of the TGF-β superfamily, in human glioma and glioblastoma cell lines. Cancer Lett 2008; 270:30-9. [DOI: 10.1016/j.canlet.2008.04.042] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 04/17/2008] [Accepted: 04/22/2008] [Indexed: 02/01/2023]
|
87
|
Yoshioka H, Kamitani H, Watanabe T, Eling TE. Nonsteroidal anti-inflammatory drug-activated gene (NAG-1/GDF15) expression is increased by the histone deacetylase inhibitor trichostatin A. J Biol Chem 2008; 283:33129-37. [PMID: 18801729 DOI: 10.1074/jbc.m805248200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Nonsteroidal anti-inflammatory drug-activated gene (NAG-1) is a putative tumor suppressor whose expression can be increased by drug treatment. Glioblastoma is the most common central nervous system tumor, is associated with high morbidity and mortality, and responds poorly to surgical, chemical, and radiation therapy. The histone deacetylase inhibitors are under current consideration as therapeutic agents in treating glioblastoma. We investigated whether trichostatin A (TSA) would alter the expression of NAG-1 in glioblastoma cells. The DNA demethylating agent 5-aza-dC did not increase NAG-1 expression, but TSA up-regulated NAG-1 expression and acted synergistically with 5-aza-dC to induce NAG-1 expression. TSA indirectly increases NAG-1 promoter activity and increases NAG-1 mRNA and protein expression in the T98G human glioblastoma cell line. TSA also increases the expression of transcription factors Sp-1 and Egr-1. Small interfering RNA experiments link NAG-1 expression to apoptosis induced by TSA. Reporter gene assays, specific inhibition by small interfering RNA transfections, and chromatin immunoprecipitation assays indicate that Egr-1 and Sp-1 mediate TSA-induced NAG-1 expression. TSA also increases the stability of NAG-1 mRNA. TSA-induced NAG-1 expression involves multiple mechanisms at the transcriptional and post-transcriptional levels.
Collapse
Affiliation(s)
- Hiroki Yoshioka
- Laboratory of Molecular Carcinogenesis, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | | | | | |
Collapse
|
88
|
Effect of in vivo loss of GDF-15 on hepatocellular carcinogenesis. J Cancer Res Clin Oncol 2008; 134:753-9. [PMID: 18210153 DOI: 10.1007/s00432-007-0336-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Accepted: 11/08/2007] [Indexed: 01/08/2023]
Abstract
BACKGROUND Growth/differentiation factor-15(GDF-15) is a divergent TGF-beta family member that is expressed following liver injury and carcinogen exposure. GDF-15 expression is highly associated with gastrointestinal cancer stage, size, and metastasis and has been implicated in inhibition of tumor growth and increased tumor invasiveness. The current study sought to determine the effect of GDF-15 ablation on the development of hepatocellular carcinoma (HCC) in vivo. MATERIALS AND METHODS Male mice genetically deleted for the gene encoding GDF-15 (Gdf15 (-/-) mice) and wild-type controls were exposed to the hepatocarcinogen diethylnitrosamine (DEN). Mice were killed at 6 months of age and their livers dissected and processed for histology. Tumor number and size relative to total liver area examined were determined. RESULTS At 6 months of age, tumors were identified in 16 of 20 (80%) Gdf15 (-/-) mice and 16 of 19 wild-type mice (84%). No significant difference in tumor-occupied area was observed in Gdf15 (-/-) mice versus wild-type mice. In addition, no difference in invasiveness was observed in HCC arising in Gdf15 (-/-) as compared to wild-type mice. In wild type mice strong immunohistochemical staining for GDF-15 was noted on small HCC foci, whereas a loss of GDF-15 expression was found in a number of advanced HCC tumors. CONCLUSIONS Although highly expressed in association with multiple gastrointestinal cancers, and lost in some advanced HCC, genetic ablation of GDF-15 has no apparent effect on HCC tumor formation rate, growth rate or invasiveness in diethylnitrosamine-induced HCC in vivo.
Collapse
|
89
|
Koike N, Kassai Y, Kouta Y, Miwa H, Konishi M, Itoh N. Brorin, a Novel Secreted Bone Morphogenetic Protein Antagonist, Promotes Neurogenesis in Mouse Neural Precursor Cells. J Biol Chem 2007; 282:15843-50. [PMID: 17400546 DOI: 10.1074/jbc.m701570200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We identified a gene encoding a novel secreted protein in mice and humans and named it Brorin. Mouse Brorin consists of 324 amino acids with a putative secreted signal sequence at its amino terminus and two cysteine-rich domains in its core region. Positions of 10 cysteine residues in the domains of Brorin are similar to those in the cysteine-rich domains of members of the Chordin family. However, the amino acid sequence of Brorin is not significantly similar to that of any other member of the Chordin family, indicating that Brorin is a unique member of the family. Mouse Brorin protein produced in cultured cells was efficiently secreted into the culture medium. The protein inhibited the activity of bone morphogenetic protein 2 (BMP2) and BMP6 in mouse preosteoblastic MC3T3-E1 cells. Mouse Brorin was predominantly expressed in neural tissues in embryos and also predominantly expressed in the adult brain. In the brain, the expression was detected in neurons, but not glial cells. The neural tissue-specific expression profile of Brorin is quite distinct from that of any other member of the Chordin family. Brorin protein promoted neurogenesis, but not astrogenesis, in mouse neural precursor cells. The present findings indicate that Brorin is a novel secreted BMP antagonist that potentially plays roles in neural development and functions.
Collapse
Affiliation(s)
- Naomi Koike
- Department of Genetic Biochemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto 606-8501, Japan
| | | | | | | | | | | |
Collapse
|
90
|
Chen SJ, Karan D, Johansson SL, Lin FF, Zeckser J, Singh AP, Batra SK, Lin MF. Prostate-derived factor as a paracrine and autocrine factor for the proliferation of androgen receptor-positive human prostate cancer cells. Prostate 2007; 67:557-71. [PMID: 17221842 DOI: 10.1002/pros.20551] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND The expression of prostate-derived factor (PDF) is significantly elevated in human prostate tumors. We investigate the functional role and signaling of PDF in androgen receptor (AR)-positive human prostate cancer cells. METHODS Transient or stable expression of PDF by cDNA transfection, antisense-mediated gene silencing, media conditioned by PDF-elevated cells, and antibody (Ab) neutralization were employed. RESULTS Elevated endogenous and exogenous expression of PDF and treatment of PDF-enriched media were associated with increased proliferation and clonogenic growth of the cells. On the contrary, knockdown of PDF or addition of PDF neutralizing Ab resulted in diminished proliferation and reduced anchorage-independent growth. Further, ERK1/2 and p90RSK, but not Smad2/3, were activated in PDF-elevated cells as well as in cells treated with PDF-enriched media, while inhibition of ERK1/2 decreased the growth of those cells. CONCLUSION PDF promotes AR-positive prostate tumor progression through upregulating cell proliferation via ERK1/2 signal pathway.
Collapse
Affiliation(s)
- Siu-Ju Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Kempf T, Horn-Wichmann R, Brabant G, Peter T, Allhoff T, Klein G, Drexler H, Johnston N, Wallentin L, Wollert KC. Circulating Concentrations of Growth-Differentiation Factor 15 in Apparently Healthy Elderly Individuals and Patients with Chronic Heart Failure as Assessed by a New Immunoradiometric Sandwich Assay. Clin Chem 2007; 53:284-91. [PMID: 17185363 DOI: 10.1373/clinchem.2006.076828] [Citation(s) in RCA: 217] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Abstract
Background: Growth-differentiation factor 15 (GDF15) is a member of the transforming growth factor β (TGF-β) cytokine superfamily. There has been increasing interest in using circulating GDF15 as a biomarker in patients, for example those with cardiovascular disease.
Methods: We developed an IRMA that uses a polyclonal, affinity chromatography–purified goat antihuman GDF15 IgG antibody, assessed the preanalytic characteristics of GDF15, and determined circulating GDF15 concentrations in 429 apparently healthy elderly individuals and 153 patients with chronic heart failure (CHF).
Results: The assay had a detection limit of 20 ng/L, an intraassay imprecision of ≤10.6%, and an interassay imprecision of ≤12.2%. Specificity was demonstrated with size-exclusion chromatography, parallel measurements with polyclonal and monoclonal anti-GDF15 antibody, and lack of cross-reactivity with TGF-β. The assay was not appreciably influenced by the anticoagulant matrix or unrelated biological substances. GDF15 was stable at room temperature for 48 h and resistant to 4 freeze-thaw cycles. Apparently healthy, elderly individuals presented with a median GDF15 concentration of 762 ng/L (25th–75th percentiles, 600–959 ng/L). GDF15 concentrations were associated with age and with cystatin C and C-reactive protein concentrations. CHF patients had increased GDF15 concentrations that were closely related to disease severity.
Conclusion: The IRMA can detect GDF15 in human serum and plasma with excellent sensitivity and specificity. The reference limits and confounding variables defined for apparently healthy elderly individuals and the favorable preanalytic characteristics of GDF15 are expected to facilitate future studies of GDF15 as a biomarker in various disease settings, including CHF.
Collapse
Affiliation(s)
- Tibor Kempf
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Gash DM, Chen Y, Gerhardt G. Neurotrophic factors and Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2007; 83:521-533. [PMID: 18808932 DOI: 10.1016/s0072-9752(07)83025-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
|
93
|
Lambert JR, Kelly JA, Shim M, Huffer WE, Nordeen SK, Baek SJ, Eling TE, Lucia MS. Prostate derived factor in human prostate cancer cells: gene induction by vitamin D via a p53-dependent mechanism and inhibition of prostate cancer cell growth. J Cell Physiol 2006; 208:566-74. [PMID: 16741990 DOI: 10.1002/jcp.20692] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The secosteroid hormone 1alpha, 25-dihydroxyvitamin D3 (1,25D) has been shown to regulate the growth and differentiation of human prostate cancer (PCa) cells, although the precise molecular mechanisms mediating these effects have not been defined. Previous studies in our laboratory demonstrated that the antiproliferative effects of 1,25D on PCa cells are mediated through the nuclear vitamin D receptor (VDR). In the present study, we performed gene profiling of LNCaP human PCa cells following 1,25D treatment and identified the antitumorigenic gene, prostate derived factor (PDF), as being highly induced by 1,25D. PDF is a member of the TGF-beta superfamily and has been implicated in a variety of functions directly related totumorigenicity including antiproliferative and pro-apoptotic effects. Gene expression studies using 1,25D analogs and a VDR antagonist demonstrate that 1,25D-mediated induction of PDF message and protein in PCa cells is dependent on VDR action. PDF is a transcriptional target of the tumor suppressor, p53. Here we show that the expression of PDF in nine PCa cell lines is dependent on functional p53. Additionally, transfection of p53-null ALVA-31 PCa cells with a p53 expression plasmid, and expression of dominant negative p53 in LNCaP PCa cells, show that the ability of VDR to induce PDF requires functional p53. Importantly, forced PDF expression in PC-3 cells results in decreased cell proliferation, soft agar cloning, and xenograft tumor size. These data demonstrate that PDF exerts antitumorigenic properties on PCa cells and its regulation by 1,25D may provide insights into the action of 1,25D in PCa.
Collapse
Affiliation(s)
- James R Lambert
- Department of Pathology, University of Colorado Health Sciences Center, Aurora, Colorado 80045, USA.
| | | | | | | | | | | | | | | |
Collapse
|
94
|
Satoh JI, Tabunoki H, Nanri Y, Arima K, Yamamura T. Human astrocytes express 14-3-3 sigma in response to oxidative and DNA-damaging stresses. Neurosci Res 2006; 56:61-72. [PMID: 16797759 DOI: 10.1016/j.neures.2006.05.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2006] [Revised: 04/17/2006] [Accepted: 05/17/2006] [Indexed: 10/24/2022]
Abstract
The 14-3-3 protein family consists of seven isoforms, most of which are expressed abundantly in neurons and glial cells, although the sigma isoform, a p53 target gene originally identified as an epithelium-specific marker, has not been identified in the human central nervous system. Here, we show that human astrocytes in culture expressed 14-3-3sigma under stress conditions. By Western blot, the expression of 14-3-3sigma, p53 and p21 was coordinately upregulated in astrocytes following exposure to hydrogen peroxide, 4-hydroxy-2-nonenal (4-HNE) or etoposide, a topoisomerase II inhibitor. 14-3-3sigma was induced by treatment with 5-aza-2'-deoxycytidine, suggesting a hypermethylated status of the gene promoter in astrocytes. In vivo, a small subset of hypertrophic reactive astrocytes, often showing a multinucleated morphology, expressed 14-3-3sigma in active demyelinating lesions of multiple sclerosis (MS) and ischemic lesions of cerebral infarction, where the expression of 4-HNE and 8-hydroxy-2'-deoxyguanosine was enhanced in reactive astrocytes. Microarray analysis of etoposide-treated astrocytes verified upregulation of p53-responsive genes and concurrent downregulation of mitotic checkpoint-regulatory genes. These observations suggest that 14-3-3sigma might serve as a marker of oxidative and DNA-damaging stresses inducing the mitotic checkpoint dysfunction in reactive astrocytes under pathological conditions.
Collapse
Affiliation(s)
- Jun-ichi Satoh
- Department of Bioinformatics and Neuroinformatics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
95
|
Boyer J, Allen WL, McLean EG, Wilson PM, McCulla A, Moore S, Longley DB, Caldas C, Johnston PG. Pharmacogenomic identification of novel determinants of response to chemotherapy in colon cancer. Cancer Res 2006; 66:2765-77. [PMID: 16510598 DOI: 10.1158/0008-5472.can-05-2693] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
DNA microarray analysis was used to analyze the transcriptional profile of HCT116 colorectal cancer cells that were treated with 5-fluorouracil (5-FU) or oxaliplatin and selected for resistance to these agents. Bioinformatic analyses identified sets of genes that were constitutively dysregulated in drug-resistant cells and transiently altered following acute exposure of parental cells to drug. We propose that these genes may represent molecular signatures of sensitivity to 5-FU and oxaliplatin. Using real-time reverse transcription-PCR (RT-PCR), the robustness of our microarray data was shown with a strong overall concordance of expression trends for > or =82% (oxaliplatin) and > or =85% (5-FU) of a representative subset of genes. Furthermore, strong correlations between the microarray and real-time RT-PCR measurements of average fold changes in gene expression were observed for both the 5-FU (R(2) > or = 0.73) and oxaliplatin gene sets (R(2) > or = 0.63). Functional analysis of three genes identified in the microarray study [prostate-derived factor (PDF), calretinin, and spermidine/spermine N(1)-acetyl transferase (SSAT)] revealed their importance as novel regulators of cytotoxic drug response. These data show the power of this novel microarray-based approach to identify genes which may be important markers of response to treatment and/or targets for therapeutic intervention.
Collapse
Affiliation(s)
- John Boyer
- Department of Oncology, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, Northern Ireland
| | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Brown DA, Stephan C, Ward RL, Law M, Hunter M, Bauskin AR, Amin J, Jung K, Diamandis EP, Hampton GM, Russell PJ, Giles GG, Breit SN. Measurement of serum levels of macrophage inhibitory cytokine 1 combined with prostate-specific antigen improves prostate cancer diagnosis. Clin Cancer Res 2006; 12:89-96. [PMID: 16397029 DOI: 10.1158/1078-0432.ccr-05-1331] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE Current serum testing for the detection of prostate cancer (PCa) lacks specificity. On diagnosis, the optimal therapeutic pathway is not clear and tools for adequate risk assessment of localized PCa progression are not available. This leads to a significant number of men having unnecessary diagnostic biopsies and surgery. A search for novel tumor markers identified macrophage inhibitory cytokine 1 (MIC-1) as a potentially useful marker. Follow-up studies revealed MIC-1 overexpression in local and metastatic PCa whereas peritumoral interstitial staining for MIC-1 identified lower-grade tumors destined for recurrence. Consequently, we sought to assess serum MIC-1 measurement as a diagnostic tool. EXPERIMENTAL DESIGN Using immunoassay determination of serum MIC-1 concentration in 1,000 men, 538 of whom had PCa, we defined the relationship of MIC-1 to disease variables. A diagnostic algorithm (MIC-PSA score) based on serum levels of MIC-1, total serum prostate-specific antigen, and percentage of free prostate-specific antigen was developed. RESULTS Serum MIC-1 was found to be an independent predictor of the presence of PCa and tumors with a Gleason sum > or =7. We validated the MIC-PSA score in a separate population and showed an improved specificity for diagnostic blood testing for PCa over percentage of free prostate-specific antigen, potentially reducing unnecessary biopsies by 27%. CONCLUSIONS Serum MIC-1 is an independent marker of the presence of PCa and tumors with a Gleason sum of > or =7. The use of serum MIC-1 significantly increases diagnostic specificity and may be a future tool in the management of PCa.
Collapse
Affiliation(s)
- David A Brown
- Centre for Immunology at St. Vincent's Hospital, Sydney, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Zimmers TA, Jin X, Hsiao EC, Perez EA, Pierce RH, Chavin KD, Koniaris LG. Growth Differentiation Factor-15: Induction in Liver Injury Through p53 and Tumor Necrosis Factor-Independent Mechanisms1. J Surg Res 2006; 130:45-51. [PMID: 16154591 DOI: 10.1016/j.jss.2005.07.036] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2005] [Revised: 06/04/2005] [Accepted: 07/23/2005] [Indexed: 01/16/2023]
Abstract
Expression of macrophage inhibitory cytokine-1 (MIC-1), a divergent transforming growth factor-beta family member, and its murine ortholog, growth/differentiation factor-15 (GDF-15), is induced in hepatocytes by surgical and chemical injury and heat shock. Here, we demonstrate that the regulation of GDF-15/MIC-1 expression may be evolutionarily conserved because MIC-1 was induced in diseased human livers. Gdf15 induction was independent of protein synthesis, a hallmark of immediate-early gene regulation. Although tumor necrosis factor (TNF) induced GDF-15 expression, injury-elicited Gdf15 expression was not reduced in mice deficient for both TNF receptor subtypes. Furthermore, although the stress sensor p53 is known to induce GDF-15/MIC-1 expression, injury-elicited Gdf15 expression was unchanged in p53 null mice. Our results demonstrate that GDF-15 induction is an immediate early response to liver injury that can occur through TNF and p53 independent pathways.
Collapse
Affiliation(s)
- Teresa A Zimmers
- DeWitt Daughtry Family Department of Surgery and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | | | | | | | | | | | | |
Collapse
|
98
|
Redzic ZB, Preston JE, Duncan JA, Chodobski A, Szmydynger-Chodobska J. The Choroid Plexus‐Cerebrospinal Fluid System: From Development to Aging. Curr Top Dev Biol 2005; 71:1-52. [PMID: 16344101 DOI: 10.1016/s0070-2153(05)71001-2] [Citation(s) in RCA: 225] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The function of the cerebrospinal fluid (CSF) and the tissue that secretes it, the choroid plexus (CP), has traditionally been thought of as both providing physical protection to the brain through buoyancy and facilitating the removal of brain metabolites through the bulk drainage of CSF. More recent studies suggest, however, that the CP-CSF system plays a much more active role in the development, homeostasis, and repair of the central nervous system (CNS). The highly specialized choroidal tissue synthesizes trophic and angiogenic factors, chemorepellents, and carrier proteins, and is strategically positioned within the ventricular cavities to supply the CNS with these biologically active substances. Through polarized transport systems and receptor-mediated transcytosis across the choroidal epithelium, the CP, a part of the blood-CSF barrier (BCSFB), controls the entry of nutrients, such as amino acids and nucleosides, and peptide hormones, such as leptin and prolactin, from the periphery into the brain. The CP also plays an important role in the clearance of toxins and drugs. During CNS development, CP-derived growth factors, such as members of the transforming growth factor-beta superfamily and retinoic acid, play an important role in controlling the patterning of neuronal differentiation in various brain regions. In the adult CNS, the CP appears to be critically involved in neuronal repair processes and the restoration of the brain microenvironment after traumatic and ischemic brain injury. Furthermore, recent studies suggest that the CP acts as a nursery for neuronal and astrocytic progenitor cells. The advancement of our knowledge of the neuroprotective capabilities of the CP may therefore facilitate the development of novel therapies for ischemic stroke and traumatic brain injury. In the later stages of life, the CP-CSF axis shows a decline in all aspects of its function, including CSF secretion and protein synthesis, which may in themselves increase the risk for development of late-life diseases, such as normal pressure hydrocephalus and Alzheimer's disease. The understanding of the mechanisms that underlie the dysfunction of the CP-CSF system in the elderly may help discover the treatments needed to reverse the negative effects of aging that lead to global CNS failure.
Collapse
Affiliation(s)
- Zoran B Redzic
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD United Kingdom
| | | | | | | | | |
Collapse
|
99
|
Harvey BK, Hoffer BJ, Wang Y. Stroke and TGF-beta proteins: glial cell line-derived neurotrophic factor and bone morphogenetic protein. Pharmacol Ther 2004; 105:113-25. [PMID: 15670622 DOI: 10.1016/j.pharmthera.2004.09.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2004] [Accepted: 09/24/2004] [Indexed: 10/26/2022]
Abstract
Recent studies have indicated that proteins in the transforming growth factor-beta superfamily alter damage induced by various neuronal injuries. Of these proteins, glial cell line-derived neurotrophic factor (GDNF) and bone morphogenetic protein-7 (BMP-7) have unique protective and regenerative effects in stroke animals. Delivery of GDNF or BMP-7 to brain tissue reduced cerebral infarction and improved motor functions in stroke animals. Pretreatment with these factors reduced caspase-3 activity and DNA fragmentation in the ischemic brain region, suggesting that antiapoptotic effects are involved. Beside the protective effects, BMP-7 given after stroke improves locomotor function. These regenerative effects of BMP-7 may involve the enhancement of dendritic growth and remodeling. In this review, we illustrate the neuroprotective and neuroregenerative properties of GDNF and BMP-7 and emphasize their therapeutic potential for stroke.
Collapse
Affiliation(s)
- Brandon K Harvey
- Neural Protection and Regeneration Section, Molecular Neuropsychiatry Branch, National Institute on Drug Abuse, NIH, Baltimore, MD 21124, USA
| | | | | |
Collapse
|
100
|
O'Keeffe GW, Dockery P, Sullivan AM. Effects of growth/differentiation factor 5 on the survival and morphology of embryonic rat midbrain dopaminergic neurones in vitro. ACTA ACUST UNITED AC 2004; 33:479-88. [PMID: 15906156 DOI: 10.1007/s11068-004-0511-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Revised: 08/11/2004] [Accepted: 08/18/2004] [Indexed: 12/24/2022]
Abstract
Growth/differentiation factor 5 (GDF5) is a member of the transforming growth factor-beta superfamily that is expressed in the developing CNS, including the ventral mesencephalon (VM). GDF5 has been shown to increase the survival of dopaminergic neurones in animal models of Parkinson's disease. This study was aimed at characterising the effects of GDF5 on dopaminergic neurones in vitro. Treatment with GDF5 induced a three-fold increase in the number of dopaminergic neurones in embryonic day 14 rat VM cultures after six days in vitro. A significant increase was also observed in the numbers of astrocytes in GDF5-treated cultures. GDF5 treatment also had significant effects on the morphology of dopaminergic neurones in these cultures; total neurite length, number of branch points and somal area were all significantly increased after six days in vitro. Analysis of neurite length and numbers of branch points at each level of the neuritic field revealed that the most pronounced effects of GDF5 were on the secondary and tertiary levels of the neuritic field. The specific type I receptor for GDF5, bone morphogenetic protein receptor (BMPR)-Ib, was found to be strongly expressed in freshly-dissected E14 VM tissue, but its expression was lost with increasing time in culture. Accordingly, treatment with GDF5 for 24 h from the time of plating induced increases in the numbers of dopaminergic neurones, while treatment with GDF5 for 24 h after six days in vitro did not. This study shows that GDF5 can promote both the survival and morphological differentiation of VM dopaminergic neurones in vitro, lending support to its potential as a candidate dopaminergic neurotrophin for use in the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Gerard W O'Keeffe
- Department of Neuroscience/Anatomy, Biosciences Research Institute, National University of Ireland Cork (NUIC), Cork, Ireland
| | | | | |
Collapse
|