1
|
Zhang K, Xie H, Zhao F, Huang Y. Identification of the gene signatures related to NK/T cell communication to evaluate the tumor microenvironment and prognostic outcomes of patients with prostate adenocarcinoma. Front Immunol 2025; 16:1564784. [PMID: 40308606 PMCID: PMC12041053 DOI: 10.3389/fimmu.2025.1564784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Background Prostate adenocarcinoma (PRAD) is a leading cause of male mortality, with NK/T cell communication being key areas of the research. Methods The Seurat package was utilized to normalize and reduce the dimensionality of the single-cell data, and CellMarker 2.0 was employed for cell annotation. CellChat was utilized to construct the ligand-receptor interaction network of cell subsets. Differentially expressed genes (DEGs) were filtered by the limma package. Univariate Cox and the LASSO regression in the glmnet package were used to obtain biomarkers and develop a risk model. The survminer package was used to calculate the optimal threshold for dividing patients into high-risk and low-risk groups, and then Kaplan-Meier (KM) survival analysis was performed. Single-sample GSEA (ssGSEA), TIMER, and ESTIMATE packages were employed for immune infiltration analysis. Pathway analysis was conducted for the low- and high-risk groups using GSEA. Immunotherapy responses were evaluated by adopting TIDE method. Additional cellular validation (quantitative real-time PCR, CCK-8, Transwell, and scratch assay) was implemented to confirm the effects of feature genes on PRAD. Results Compared with the benign group, NK/T cells were the cell type with the greatest changes in the tumor group, and their communication intensity was relatively high among all cell types. A RiskScore model was constructed as follows: 0.579 * F O X S 1 + 0.345 * G P C 6 + 0.385 * I S Y N A 1 + 0.418 * I T G A X + 0.792 * M G A T 4 B + 0.368 * P R R 7 + 0.458 * R E X O 2 . Analysis of the differences between the two risk groups showed that the level of immune infiltration was higher in the high-risk group, and it was significantly enriched in immune-correlated pathways, while the low-risk group was mainly enriched in metabolism-related pathways. TIDE analysis indicated that the high-risk group had higher immune escape potential. The cellular validation assays have revealed the higher expression of seven biomarkers in PRAD groups. Further, ISYNA1 knockdown inhibited the proliferation, migration, and invasion ability of PRAD cells. Conclusion The current research reveals key communication genes in PRAD, offering new possibilities for the exploration of new therapeutic targets.
Collapse
Affiliation(s)
| | | | | | - Yeqing Huang
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
2
|
Lasconi C, Pahl MC, Cousminer DL, Doege CA, Chesi A, Hodge KM, Leonard ME, Lu S, Johnson ME, Su C, Hammond RK, Pippin JA, Terry NA, Ghanem LR, Leibel RL, Wells AD, Grant SFA. Variant-to-Gene-Mapping Analyses Reveal a Role for the Hypothalamus in Genetic Susceptibility to Inflammatory Bowel Disease. Cell Mol Gastroenterol Hepatol 2020; 11:667-682. [PMID: 33069917 PMCID: PMC7843407 DOI: 10.1016/j.jcmgh.2020.10.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Inflammatory bowel disease (IBD) is a polygenic disorder characterized principally by dysregulated inflammation impacting the gastrointestinal tract. However, there also is increasing evidence for a clinical association with stress and depression. Given the role of the hypothalamus in stress responses and in the pathogenesis of depression, useful insights could be gleaned from understanding its genetic role in IBD. METHODS We conducted genetic correlation analyses on publicly available genome-wide association study summary statistics for depression and IBD traits to identify genetic commonalities. We used partitioned linkage disequilibrium score regression, leveraging our ATAC sequencing and promoter-focused Capture C data, to measure enrichment of IBD single-nucleotide polymorphisms within promoter-interacting open chromatin regions of human embryonic stem cell-derived hypothalamic-like neurons (HNs). Using the same data sets, we performed variant-to-gene mapping to implicate putative IBD effector genes in HNs. To contrast these results, we similarly analyzed 3-dimensional genomic data generated in epithelium-derived colonoids from rectal biopsy specimens from donors without pathologic disease noted at the time of colonoscopy. Finally, we conducted enrichment pathway analyses on the implicated genes to identify putative IBD dysfunctional pathways. RESULTS We found significant genetic correlations (rg) of 0.122 with an adjusted P (Padj) = 1.4 × 10-4 for IBD: rg = 0.122; Padj = 2.5 × 10-3 for ulcerative colitis and genetic correlation (rg) = 0.094; Padj = 2.5 × 10-3 for Crohn's disease, and significant approximately 4-fold (P = .005) and approximately 7-fold (P = .03) enrichment of IBD single-nucleotide polymorphisms in HNs and colonoids, respectively. We implicated 25 associated genes in HNs, among which CREM, CNTF, and RHOA encode key regulators of stress. Seven genes also additionally were implicated in the colonoids. We observed an overall enrichment for immune and hormonal signaling pathways, and a colonoid-specific enrichment for microbiota-relevant terms. CONCLUSIONS Our results suggest that the hypothalamus warrants further study in the context of IBD pathogenesis.
Collapse
Affiliation(s)
- Chiara Lasconi
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania
| | - Matthew C Pahl
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania
| | - Diana L Cousminer
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania
| | - Claudia A Doege
- Division of Molecular Genetics (Pediatrics), Naomi Berrie Diabetes Center, Columbia University Vagelos College of Physicians and Surgeons, New York, New York
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania
| | - Kenyaita M Hodge
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania
| | - Michelle E Leonard
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania
| | - Sumei Lu
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania
| | - Matthew E Johnson
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania
| | - Chun Su
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania
| | - Reza K Hammond
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania
| | - James A Pippin
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania
| | | | | | - Rudolph L Leibel
- Division of Molecular Genetics (Pediatrics), Naomi Berrie Diabetes Center, Columbia University Vagelos College of Physicians and Surgeons, New York, New York
| | - Andrew D Wells
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Department of Pathology, Philadelphia, Pennsylvania; Department of Pathology and Laboratory Medicine, Philadelphia, Pennsylvania
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, Philadelphia, Pennsylvania; Division of Human Genetics, Philadelphia, Pennsylvania; Division of Diabetes and Endocrinology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
3
|
Montoya A, Yepes L, Bedoya A, Henao R, Delgado G, Vélez ID, Robledo SM. Transforming Growth Factor Beta (TGFβ1) and Epidermal Growth Factor (EGF) as Biomarkers of Leishmania (V) braziliensis Infection and Early Therapeutic Response in Cutaneous Leishmaniasis: Studies in Hamsters. Front Cell Infect Microbiol 2018; 8:350. [PMID: 30333964 PMCID: PMC6176012 DOI: 10.3389/fcimb.2018.00350] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 09/13/2018] [Indexed: 12/19/2022] Open
Abstract
Introduction: In cutaneous leishmaniasis, the host immune response is responsible for the development of skin injuries but also for resolution of the disease especially after antileishmanial therapy. The immune factors that participate in the regulation of inflammation, remodeling of the extracellular matrix, cell proliferation and differentiation may constitute biomarkers of diseases or response to treatment. In this work, we analyzed the production of the growth factors EGF, TGFβ1, PDGF, and FGF during the infection by Leishmania parasites, the development of the injuries and the early response to treatment. Methodology: Golden hamsters were infected with L. (V) braziliensis. The growth factors were detected in skin scrapings and biopsies every 2 weeks after infected and then at day 7 of treatment with different drug candidates by RT-qPCR. The parasitic load was also quantified by RT-qPCR in skin biopsies sampled at the end of the study. Results: The infection by L. (V) braziliensis induced the expression of all the growth factors at day 15 of infection. One month after infection, EGF and TGFβ1 were expressed in all hamsters with inverse ratio. While the EGF and FGF levels decreased between day 15 and 30 of infection, the TGFβ1 increased and the PGDF levels did not change. The relative expression of EGF and TGFβ1 increased notably after treatment. However, the increase of EGF was associated with clinical cure while the increase of TGFβ1 was associated with failure to treatment. The amount of parasites in the cutaneous lesion at the end of the study decreased according to the clinical outcome, being lower in the group of cured hamsters and higher in the group of hamsters that had a failure to the treatment. Conclusions: A differential profile of growth factor expression occurred during the infection and response to treatment. Higher induction of TGFβ1 was associated with active disease while the higher levels of EGF are associated with adequate response to treatment. The inversely EGF/TGFβ1 ratio may be an effective biomarker to identify establishment of Leishmania infection and early therapeutic response, respectively. However, further studies are needed to validate the utility of the proposed biomarkers in field conditions.
Collapse
Affiliation(s)
- Andrés Montoya
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Lina Yepes
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Alexander Bedoya
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Raúl Henao
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Gabriela Delgado
- Grupo de Investigación en Inmunotoxicología, Departamento de Farmacia, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Iván D Vélez
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Sara M Robledo
- Programa de Estudio y Control de Enfermedades Tropicales (PECET), Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
4
|
Reciprocal control of excitatory synapse numbers by Wnt and Wnt inhibitor PRR7 secreted on exosomes. Nat Commun 2018; 9:3434. [PMID: 30143647 PMCID: PMC6109165 DOI: 10.1038/s41467-018-05858-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 07/25/2018] [Indexed: 12/22/2022] Open
Abstract
Secreted Wnts play crucial roles in synaptogenesis and synapse maintenance, but endogenous factors promoting synapse elimination in central neurons remain unknown. Here we show that proline-rich 7 (PRR7) induces specific removal of excitatory synapses and acts as a Wnt inhibitor. Remarkably, transmembrane protein PRR7 is activity-dependently released by neurons via exosomes. Exosomal PRR7 is uptaken by neurons through membrane fusion and eliminates excitatory synapses in neighboring neurons. Conversely, PRR7 knockdown in sparse neurons greatly increases excitatory synapse numbers in all surrounding neurons. These non-cell autonomous effects of PRR7 are effectively negated by augmentation or blockade of Wnt signaling. PRR7 exerts its effect by blocking the exosomal secretion of Wnts, activation of GSK3β, and promoting proteasomal degradation of PSD proteins. These data uncover a proximity-dependent, reciprocal mechanism for the regulation of excitatory synapse numbers in local neurons and demonstrate the significance of exosomes in inter-neuronal signaling in the vertebrate brain. Wnts are important for synapse formation and maintenance. Here, the authors show that proline-rich 7 (PRR7) is a Wnt inhibitor that is secreted via exosomes to regulate excitatory synapse numbers.
Collapse
|
5
|
Curson JEB, Luo L, Sweet MJ, Stow JL. pTRAPs: Transmembrane adaptors in innate immune signaling. J Leukoc Biol 2018; 103:1011-1019. [PMID: 29601097 DOI: 10.1002/jlb.2ri1117-474r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/08/2018] [Accepted: 02/10/2018] [Indexed: 01/30/2023] Open
Abstract
Transmembrane adaptor proteins (TRAPs) are protein scaffolds and signaling regulators with established roles in signal-induced activation of lymphocytes. A subset of the TRAP family, the palmitoylated TRAPs (pTRAPs), are increasingly emerging with additional roles in innate immune cells. Targeted to lipid rafts, tetraspannin-enriched microdomains, and protein microclusters in membranes, pTRAP scaffolds exert spatiotemporal regulation by recruiting signaling kinases, particularly Src and Syk family members, as well as Csk, and other effectors. In this way, pTRAPs modulate signaling and influence resulting cell responses, including the selective output of inflammatory cytokines and other mediators. Here, we review studies revealing that different pTRAPs work together, often with overlapping or redundant roles, for positive and negative regulation of key innate immune pathways, including Fc receptor and pattern recognition receptor signaling. Recent findings show that pTRAPs can bind directly to innate immune receptors, in addition to other transmembrane binding partners. Thus, pTRAPs are important, multifunctional scaffolds in pathways that are fundamental to diverse innate immune responses.
Collapse
Affiliation(s)
- James E B Curson
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Lin Luo
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Jennifer L Stow
- Institute for Molecular Bioscience (IMB), IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
6
|
Kim HT, Lee MS, Jeong YM, Ro H, Kim DI, Shin YH, Kim JE, Hwang KS, Choi JH, Bahn M, Lee JJ, Lee SH, Bae YK, Lee JS, Choi JK, Kim NS, Yeo CY, Kim CH. Ottogi Inhibits Wnt/β-catenin Signaling by Regulating Cell Membrane Trafficking of Frizzled8. Sci Rep 2017; 7:13278. [PMID: 29038508 PMCID: PMC5643531 DOI: 10.1038/s41598-017-13429-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 09/22/2017] [Indexed: 01/30/2023] Open
Abstract
Wnt signaling controls critical developmental processes including tissue/body patterning. Here we report the identification of a novel regulator of Wnt signaling, OTTOGI (OTG), isolated from a large-scale expression screening of human cDNAs in zebrafish embryos. Overexpression of OTG in zebrafish embryos caused dorso-anteriorized phenotype, inhibited the expression of Wnt target genes, and prevented nuclear accumulation of β-catenin. Conversely, knockdown of zebrafish otg using specific antisense morpholino promoted nuclear accumulation of β-catenin and caused ventralization. However, OTG failed to rescue headless-like phenotype induced by inhibition of GSK-3β activity, suggesting that OTG acts upstream of GSK-3β. OTG bound specifically to Frizzled8 (Fz8) receptor and caused retention of Fz8 in the endoplasmic reticulum possibly by preventing N-linked glycosylation of Fz8. Taken together, our data indicate that OTG functions as a novel negative regulator of Wnt signaling during development by the modulation of cell surface expression of Fz receptor.
Collapse
Affiliation(s)
- Hyun-Taek Kim
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Mi-Sun Lee
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Yun-Mi Jeong
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Hyunju Ro
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Dong-Il Kim
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Yong-Hwan Shin
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Ji-Eun Kim
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Kyu-Seok Hwang
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Jung-Hwa Choi
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea
| | - Minjin Bahn
- Department of Life Science and Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 120-750, South Korea
| | - Jeong-Ju Lee
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, South Korea
| | - Sang H Lee
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Young-Ki Bae
- National Cancer Center, Goyang, 410-769, South Korea
| | - Jin-Soo Lee
- National Cancer Center, Goyang, 410-769, South Korea
| | - Joong-Kook Choi
- Department of Biochemistry, College of Medicine, Chungbuk National University, Cheongju, 361-763, South Korea
| | - Nam-Soon Kim
- Genome Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 305-806, South Korea.
| | - Chang-Yeol Yeo
- Department of Life Science and Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, 120-750, South Korea.
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, 34134, South Korea.
| |
Collapse
|
7
|
Hrdinka M, Sudan K, Just S, Drobek A, Stepanek O, Schlüter D, Reinhold D, Jordan BA, Gintschel P, Schraven B, Kreutz MR. Normal Development and Function of T Cells in Proline Rich 7 (Prr7) Deficient Mice. PLoS One 2016; 11:e0162863. [PMID: 27657535 PMCID: PMC5033326 DOI: 10.1371/journal.pone.0162863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/30/2016] [Indexed: 11/18/2022] Open
Abstract
Transmembrane adaptor proteins (TRAPs) are important organisers for the transduction of immunoreceptor-mediated signals. Prr7 is a TRAP that regulates T cell receptor (TCR) signalling and potently induces cell death when overexpressed in human Jurkat T cells. Whether endogenous Prr7 has a similar functional role is currently unknown. To address this issue, we analysed the development and function of the immune system in Prr7 knockout mice. We found that loss of Prr7 partially impairs development of single positive CD4+ T cells in the thymus but has no effect on the development of other T cell subpopulations, B cells, NK cells, or NKT cells. Moreover, Prr7 does not affect the TCR signalling pathway as T cells derived from Prr7 knockout and wild-type animals and stimulated in vitro express the same levels of the activation marker CD69, and retain their ability to proliferate and activate induced cell death programs. Importantly, Prr7 knockout mice retained the capacity to mount a protective immune response when challenged with Listeria monocytogenes infection in vivo. In addition, T cell effector functions (activation, migration, and reactivation) were normal following induction of experimental autoimmune encephalomyelitis (EAE) in Prr7 knockout mice. Collectively, our work shows that loss of Prr7 does not result in a major immune system phenotype and suggests that Prr7 has a dispensable function for TCR signalling.
Collapse
Affiliation(s)
- Matous Hrdinka
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg, Germany
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Magdeburg, Germany
- * E-mail: (MH); (BS); (MRK)
| | - Kritika Sudan
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg, Germany
| | - Sissy Just
- Institute of Medical Microbiology, Otto-von-Guericke University, Germany
| | - Ales Drobek
- Group of Adaptive Immunity, Institute of Molecular Genetics, CAS, Prague, Czech Republic
| | - Ondrej Stepanek
- Group of Adaptive Immunity, Institute of Molecular Genetics, CAS, Prague, Czech Republic
| | - Dirk Schlüter
- Institute of Medical Microbiology, Otto-von-Guericke University, Germany
- Organ-Specific Immune Regulation, Helmholtz-Center of Infection Research (HZI). Braunschweig, Germany
| | - Dirk Reinhold
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | - Bryen A. Jordan
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Patricia Gintschel
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Magdeburg, Germany
- Department of Immune Control, Helmholtz-Center of Infection Research (HZI). Braunschweig, Germany
- * E-mail: (MH); (BS); (MRK)
| | - Michael R. Kreutz
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg, Germany
- Leibniz Group 'Dendritic Organelles and Synaptic Function', University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology, Hamburg, Germany
- * E-mail: (MH); (BS); (MRK)
| |
Collapse
|
8
|
Kravchick DO, Karpova A, Hrdinka M, Lopez-Rojas J, Iacobas S, Carbonell AU, Iacobas DA, Kreutz MR, Jordan BA. Synaptonuclear messenger PRR7 inhibits c-Jun ubiquitination and regulates NMDA-mediated excitotoxicity. EMBO J 2016; 35:1923-34. [PMID: 27458189 DOI: 10.15252/embj.201593070] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 06/21/2016] [Indexed: 12/14/2022] Open
Abstract
Elevated c-Jun levels result in apoptosis and are evident in neurodegenerative disorders such as Alzheimer's disease and dementia and after global cerebral insults including stroke and epilepsy. NMDA receptor (NMDAR) antagonists block c-Jun upregulation and prevent neuronal cell death following excitotoxic insults. However, the molecular mechanisms regulating c-Jun abundance in neurons are poorly understood. Here, we show that the synaptic component Proline rich 7 (PRR7) accumulates in the nucleus of hippocampal neurons following NMDAR activity. We find that PRR7 inhibits the ubiquitination of c-Jun by E3 ligase SCF(FBW) (7) (FBW7), increases c-Jun-dependent transcriptional activity, and promotes neuronal death. Microarray assays show that PRR7 abundance is directly correlated with transcripts associated with cellular viability. Moreover, PRR7 knockdown attenuates NMDAR-mediated excitotoxicity in neuronal cultures in a c-Jun-dependent manner. Our results show that PRR7 links NMDAR activity to c-Jun function and provide new insights into the molecular processes that underlie NMDAR-dependent excitotoxicity.
Collapse
Affiliation(s)
- Dana O Kravchick
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anna Karpova
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Matous Hrdinka
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Jeffrey Lopez-Rojas
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Sanda Iacobas
- Department of Pathology, New York Medical College, Valhalla, NY, USA
| | - Abigail U Carbonell
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Dumitru A Iacobas
- Department of Pathology, New York Medical College, Valhalla, NY, USA
| | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany Leibniz Group "Dendritic Organelles and Synaptic Function", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bryen A Jordan
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
9
|
Palmitoylated transmembrane adaptor proteins in leukocyte signaling. Cell Signal 2014; 26:895-902. [PMID: 24440308 DOI: 10.1016/j.cellsig.2014.01.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/07/2014] [Accepted: 01/09/2014] [Indexed: 12/14/2022]
Abstract
Transmembrane adaptor proteins (TRAPs) are structurally related proteins that have no enzymatic function, but enable inducible recruitment of effector molecules to the plasma membrane, usually in a phosphorylation dependent manner. Numerous surface receptors employ TRAPs for either propagation or negative regulation of the signal transduction. Several TRAPs (LAT, NTAL, PAG, LIME, PRR7, SCIMP, LST1/A, and putatively GAPT) are known to be palmitoylated that could facilitate their localization in lipid rafts or tetraspanin enriched microdomains. This review summarizes expression patterns, binding partners, signaling pathways, and biological functions of particular palmitoylated TRAPs with an emphasis on the three most recently discovered members, PRR7, SCIMP, and LST1/A. Moreover, we discuss in silico methodology used for discovery of new family members, nature of their binding partners, and microdomain localization.
Collapse
|
10
|
Hrdinka M, Otahal P, Horejsi V. The transmembrane region is responsible for targeting of adaptor protein LAX into "heavy rafts". PLoS One 2012; 7:e36330. [PMID: 22662118 PMCID: PMC3360738 DOI: 10.1371/journal.pone.0036330] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 04/04/2012] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The importance of membrane compartmentalization into specific membrane microdomains has been shown in many biological processes such as immunoreceptor signaling, membrane trafficking, pathogen infection, and tumor progression. Microdomains like lipid rafts, caveolae and tetraspanin enriched microdomains are relatively resistant to solubilization by some detergents. Large detergent-resistant membrane fragments (DRMs) resulting from such membrane solubilization can be conveniently isolated by density gradient ultracentrifugation or gel filtration. Recently, we described a novel type of raft-like membrane microdomains producing, upon detergent Brij98 solubilization, "heavy DRMs" and containing a number of functionally relevant proteins. Transmembrane adaptor protein LAX is a typical "heavy raft" protein. The present study was designed to identify the molecular determinants targeting LAX-derived constructs to heavy rafts. METHODOLOGY/PRINCIPAL FINDINGS We prepared several constructs encoding chimeric proteins containing various informative segments of the LAX sequence and evaluated their effects on targeting to heavy rafts. Replacement of the polybasic membrane-proximal part of LAX by CD3ε-derived membrane-proximal part had no effect on LAX solubilization. Similarly, the membrane-proximal part of LAX, when introduced into non-raft protein CD25 did not change CD25 detergent solubility. These results indicated that membrane-proximal part of LAX is not important for LAX targeting to heavy rafts. On the other hand, the replacement of transmembrane part of CD25 by the transmembrane part of LAX resulted in targeting into heavy rafts. We also show that LAX is not S-acylated, thus palmitoylation is not involved in LAX targeting to heavy rafts. Also, covalent dimerization was excluded as a cause of targeting into heavy rafts. CONCLUSIONS/SIGNIFICANCE We identified the transmembrane domain of LAX as a first motif targeting transmembrane protein constructs to detergent-resistant heavy rafts, a novel type of membrane microdomains containing a number of physiologically important proteins.
Collapse
Affiliation(s)
- Matous Hrdinka
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Pavel Otahal
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Vaclav Horejsi
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- * E-mail:
| |
Collapse
|
11
|
Draber P, Stepanek O, Hrdinka M, Drobek A, Chmatal L, Mala L, Ormsby T, Angelisova P, Horejsi V, Brdicka T. LST1/A is a myeloid leukocyte-specific transmembrane adaptor protein recruiting protein tyrosine phosphatases SHP-1 and SHP-2 to the plasma membrane. J Biol Chem 2012; 287:22812-21. [PMID: 22589543 DOI: 10.1074/jbc.m112.339143] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transmembrane adaptor proteins are membrane-anchored proteins consisting of a short extracellular part, a transmembrane domain, and a cytoplasmic part with various protein-protein interaction motifs but lacking any enzymatic activity. They participate in the regulation of various signaling pathways by recruiting other proteins to the proximity of cellular membranes where the signaling is often initiated and propagated. In this work, we show that LST1/A, an incompletely characterized protein encoded by MHCIII locus, is a palmitoylated transmembrane adaptor protein. It is expressed specifically in leukocytes of the myeloid lineage, where it localizes to the tetraspanin-enriched microdomains. In addition, it binds SHP-1 and SHP-2 phosphatases in a phosphotyrosine-dependent manner, facilitating their recruitment to the plasma membrane. These data suggest a role for LST1/A in negative regulation of signal propagation.
Collapse
Affiliation(s)
- Peter Draber
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, 142 20 Prague, Czech Republic
| | | | | | | | | | | | | | | | | | | |
Collapse
|