1
|
Wei C, Zhang G, Fu X, Zhao M, Zhai W, Shen Y, Sun L. Correlation of peripheral olfactomedin 1 with Alzheimer's disease and cognitive functions. Transl Psychiatry 2025; 15:146. [PMID: 40221403 PMCID: PMC11993663 DOI: 10.1038/s41398-025-03373-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 12/05/2024] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
Olfactomedin 1 (OLFM1) is thought to be involved in neuronal development, synaptic structure and function. However, the expression level of peripheral OLFM1 in Alzheimer's disease (AD) and its role in AD are unclear. The present study was conducted to assess the relationship of serum OLFM1 with AD and cognitive function. This study comprised 120 patients with AD and 118 healthy controls (HC). Serum OLFM1 levels, cognitive functions, and brain region volumes were evaluated in all participants. The results demonstrated a significant reduction in serum OLFM1 levels in AD patients (749.8 ± 42.3 pg/mL) compared to HC (804.4 ± 45.7 pg/mL). Among participants carrying the APOE ε4 allele, a significant positive correlation was observed between OLFM1 levels and cognitive assessments, including Mini-Mental State Examination (MMSE), Montreal Cognitive Assessment (MoCA), and Memory and Executive Screening (MES). Furthermore, reduced OLFM1 levels were significantly associated with hippocampus (β = 0.005, 95% CI = 0.001-0.011, p = 0.042) and angular gyrus (β = 0.012, 95% CI = 0.001-0.022, p = 0.025) atrophy. The integration of serum OLFM1 with basic clinical characteristics exhibited robust discriminatory power in differentiating AD patients from HC, evidenced by an area under the curve of 0.881 (95% CI = 0.834-0.926). In summary, serum OLFM1 is a potential peripheral biomarker for AD, that correlates with cognitive function and specific brain volumes. In addition, APOE ε4 may modulate the influence of OLFM1 on cognitive function.
Collapse
Affiliation(s)
- Chunxiao Wei
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Guimei Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xiaoshu Fu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Meng Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Weijie Zhai
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yanxin Shen
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China.
- Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
2
|
Oliveira-Valença VM, Roberts JM, Fernandes-Cerqueira VM, Colmerauer CH, de Toledo BC, Santos-França PL, Linden R, Martins RAP, Rocha-Martins M, Bosco A, Vetter ML, da Silveira MS. POU4F2 overexpression promotes the genesis of retinal ganglion cell-like projection neurons from late progenitors. Development 2025; 152:DEV204297. [PMID: 39946314 DOI: 10.1242/dev.204297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 02/03/2025] [Indexed: 03/04/2025]
Abstract
Retinal ganglion cells (RGCs) are the projection neurons of the retina, and their death promotes an irreversible blindness. Several factors were described to control their genesis during retinal development. These include Atoh7, a major orchestrator of the RGC program, and downstream targets of this transcription factor, including Pou4f factors, that in turn regulate key aspects of terminal differentiation. The absence of POU4F family genes results in defects in RGC differentiation, aberrant axonal elaboration and, ultimately, RGC death. This confirms the requirement of POU4F factors for RGC development and survival, with a crucial role in regulating RGC axon outgrowth and pathfinding. Here, we have investigated in vivo whether ectopic Pou4f2 expression in late retinal progenitor cells (late RPCs) is sufficient to induce the generation of cells with RGC properties, including long-range axon projections. We show that Pou4f2 overexpression generates RGC-like cells that share morphological and transcriptional features with RGCs that are normally generated during early development and extend axonal projections up to the brain. In conclusion, these results show that POU4F2 alone is sufficient to promote the crucial properties of projection neurons that arise from retinal progenitors outside their developmental window.
Collapse
Affiliation(s)
- Viviane Medeiros Oliveira-Valença
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Jacqueline Marie Roberts
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Vitória Melo Fernandes-Cerqueira
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Carolina Herkenhoff Colmerauer
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Beatriz Cardoso de Toledo
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Pedro Lucas Santos-França
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Rafael Linden
- Neurogenesis Lab, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Rodrigo Alves Portela Martins
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Maurício Rocha-Martins
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Alejandra Bosco
- Department of Neurobiology, University of Utah, School of Medicine, Salt Lake City, UT 84112, USA
| | - Monica Lynn Vetter
- Department of Neurobiology, University of Utah, School of Medicine, Salt Lake City, UT 84112, USA
| | - Mariana Souza da Silveira
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Hutchings C, Nuriel Y, Lazar D, Kohl A, Muir E, Genin O, Cinnamon Y, Benyamini H, Nevo Y, Sela-Donenfeld D. Hindbrain boundaries as niches of neural progenitor and stem cells regulated by the extracellular matrix proteoglycan chondroitin sulphate. Development 2024; 151:dev201934. [PMID: 38251863 PMCID: PMC10911165 DOI: 10.1242/dev.201934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 01/12/2024] [Indexed: 01/23/2024]
Abstract
The interplay between neural progenitors and stem cells (NPSCs), and their extracellular matrix (ECM) is a crucial regulatory mechanism that determines their behavior. Nonetheless, how the ECM dictates the state of NPSCs remains elusive. The hindbrain is valuable to examine this relationship, as cells in the ventricular surface of hindbrain boundaries (HBs), which arise between any two neighboring rhombomeres, express the NPSC marker Sox2, while being surrounded with the membrane-bound ECM molecule chondroitin sulphate proteoglycan (CSPG), in chick and mouse embryos. CSPG expression was used to isolate HB Sox2+ cells for RNA-sequencing, revealing their distinguished molecular properties as typical NPSCs, which express known and newly identified genes relating to stem cells, cancer, the matrisome and cell cycle. In contrast, the CSPG- non-HB cells, displayed clear neural-differentiation transcriptome. To address whether CSPG is significant for hindbrain development, its expression was manipulated in vivo and in vitro. CSPG manipulations shifted the stem versus differentiation state of HB cells, evident by their behavior and altered gene expression. These results provide further understanding of the uniqueness of hindbrain boundaries as repetitive pools of NPSCs in-between the rapidly growing rhombomeres, which rely on their microenvironment to maintain their undifferentiated state during development.
Collapse
Affiliation(s)
- Carmel Hutchings
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agricultural, Food, and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Yarden Nuriel
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agricultural, Food, and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Daniel Lazar
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agricultural, Food, and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agricultural, Food, and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Elizabeth Muir
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 1TN, UK
| | - Olga Genin
- Agricultural Research Organization, Volcani Center, Department of Poultry and Aquaculture Science, Rishon LeTsiyon 7505101, Israel
| | - Yuval Cinnamon
- Agricultural Research Organization, Volcani Center, Department of Poultry and Aquaculture Science, Rishon LeTsiyon 7505101, Israel
| | - Hadar Benyamini
- Info-CORE, Bioinformatics Unit of the I-CORE at the Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Yuval Nevo
- Info-CORE, Bioinformatics Unit of the I-CORE at the Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agricultural, Food, and Environmental Sciences, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| |
Collapse
|
4
|
Wang W, Ma L, Zhao Y, Liu M, Ye W, Li X. Research progress on the role of the Wnt signaling pathway in pituitary adenoma. Front Endocrinol (Lausanne) 2023; 14:1216817. [PMID: 37780610 PMCID: PMC10538627 DOI: 10.3389/fendo.2023.1216817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/28/2023] [Indexed: 10/03/2023] Open
Abstract
Pituitary adenoma (PA) is the third most common central nervous system tumor originating from the anterior pituitary, but its pathogenesis remains unclear. The Wnt signaling pathway is a conserved pathway involved in cell proliferation, Self-renewal of stem cells, and cell differentiation. It is related to the occurrence of various tumors, including PA. This article reviews the latest developments in Wnt pathway inhibitors and pathway-targeted drugs. It discusses the possibility of combining Wnt pathway inhibitors with immunotherapy to provide a theoretical basis for the combined treatment of PA.
Collapse
Affiliation(s)
| | | | | | | | | | - Xianfeng Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
5
|
Xu K, Zheng P, Zhao S, Wang J, Feng J, Ren Y, Zhong Q, Zhang H, Chen X, Chen J, Xie P. LRFN5 and OLFM4 as novel potential biomarkers for major depressive disorder: a pilot study. Transl Psychiatry 2023; 13:188. [PMID: 37280213 DOI: 10.1038/s41398-023-02490-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/20/2023] [Accepted: 05/26/2023] [Indexed: 06/08/2023] Open
Abstract
Evidences have shown that both LRFN5 and OLFM4 can regulate neural development and synaptic function. Recent genome-wide association studies on major depressive disorder (MDD) have implicated LRFN5 and OLFM4, but their expressions and roles in MDD are still completely unclear. Here, we examined serum concentrations of LRFN5 and OLFM4 in 99 drug-naive MDD patients, 90 drug-treatment MDD patients, and 81 healthy controls (HCs) using ELISA methods. The results showed that both LRFN5 and OLFM4 levels were considerably higher in MDD patients compared to HCs, and were significantly lower in drug-treatment MDD patients than in drug-naive MDD patients. However, there were no significant differences between MDD patients who received a single antidepressant and a combination of antidepressants. Pearson correlation analysis showed that they were associated with the clinical data, including Hamilton Depression Scale score, age, duration of illness, fasting blood glucose, serum lipids, and hepatic, renal, or thyroid function. Moreover, these two molecules both yielded fairly excellent diagnostic performance in diagnosing MDD. In addition, a combination of LRFN5 and OLFM4 demonstrated a better diagnostic effectiveness, with an area under curve of 0.974 in the training set and 0.975 in the testing set. Taken together, our data suggest that LRFN5 and OLFM4 may be implicated in the pathophysiology of MDD and the combination of LRFN5 and OLFM4 may offer a diagnostic biomarker panel for MDD.
Collapse
Affiliation(s)
- Ke Xu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuang Zhao
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Jiubing Wang
- Department of Clinical Laboratory, Chongqing Mental Health Centre, Chongqing, China
| | - Jinzhou Feng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Ren
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qi Zhong
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Hanping Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangyu Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianjun Chen
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- National Health Commission Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
6
|
Iyer S, Dhiman N, Zade SP, Mukherjee S, Singla N, Kumar M. Exposure to Tetrabutylammonium Bromide Impairs Cranial Neural Crest Specification, Neurogenic Program, and Brain Morphogenesis. ACS Chem Neurosci 2023; 14:1785-1798. [PMID: 37125651 DOI: 10.1021/acschemneuro.2c00728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Tetrabutylammonium bromide (TBAB) is a widely used industrial reagent and is commonly found in our aquatic ecosystem as an industrial byproduct. In humans, the ingestion of TBAB causes severe neurological impairments and disorders such as vertigo, hallucinations, and delirium. Yet, the extent of environmental risk and TBAB toxicity to human health is poorly understood. In this study, we aim to determine the developmental toxicity of TBAB using zebrafish embryos as a model and provide novel insights into the mechanism of action of such chemicals on neurodevelopment and the overall embryonic program. Our results show that exposure to TBAB results in impaired development of the brain, inner ear, and pharyngeal skeletal elements in the zebrafish embryo. TBAB treatment resulted in aberrations in the specification of the neural crest precursors, hindbrain segmentation, and otic neurogenesis. TBAB treatment also induced a surge in apoptosis in the head, tail, and trunk regions of the developing embryo. Long-term TBAB exposure resulted in cardiac edema and craniofacial defects. Further, in silico molecular docking analysis indicated that TBAB binds to AMPA receptors and modulates neural developmental genes such as olfactomedin and acetylcholinesterase in the embryonic brain. To summarize, our study highlights the novel effects of TBAB on embryonic brain formation and segmentation, ear morphogenesis, and craniofacial skeletal development.
Collapse
Affiliation(s)
- Sharada Iyer
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Uppal Road, Habsiguda, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Neha Dhiman
- Department of Biochemistry, Panjab University, Chandigarh160014, India
| | - Suraj P Zade
- Global Product Compliance─India, 301, Samved Sankul, Near MLA Hostel, Civil Lines, Nagpur 440001, India
| | - Sulagna Mukherjee
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Uppal Road, Habsiguda, Hyderabad 500007, India
| | - Neha Singla
- Department of Biophysics, Panjab University, Chandigarh160014, India
| | - Megha Kumar
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Uppal Road, Habsiguda, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
7
|
Jaitner S, Pretzsch E, Neumann J, Schäffauer A, Schiemann M, Angele M, Kumbrink J, Schwitalla S, Greten FR, Brandl L, Klauschen F, Horst D, Kirchner T, Jung A. Olfactomedin 4 associates with expression of differentiation markers but not with properties of cancer stemness, EMT nor metastatic spread in colorectal cancer. J Pathol Clin Res 2023; 9:73-85. [PMID: 36349502 PMCID: PMC9732686 DOI: 10.1002/cjp2.300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/06/2022] [Accepted: 10/13/2022] [Indexed: 11/11/2022]
Abstract
Tumor stem cells play a pivotal role in carcinogenesis and metastatic spread in colorectal cancer (CRC). Olfactomedin 4 (OLFM4) is co-expressed with the established stem cell marker leucine-rich repeat-containing G protein-coupled receptor 5 at the bottom of intestinal crypts and has been suggested as a surrogate for cancer stemness and a biomarker in gastrointestinal tumors associated with prognosis. Therefore, it was the aim of the present study to clarify whether OLFM4 is involved in carcinogenesis and metastatic spread in CRC. We used a combined approach of functional assays using forced OLFM4 overexpression in human CRC cell lines, xenograft mice, and an immunohistochemical approach using patient tissues to investigate the impact of OLFM4 on stemness, canonical Wnt signaling, properties of metastasis and differentiation as well as prognosis. OLFM4 expression correlated weakly with tumor grade in one patient cohort (metastasis collection: p = 0.05; pooled analysis of metastasis collection and survival collection: p = 0.19) and paralleled the expression of differentiation markers (FABP2, MUC2, and CK20) (p = 0.002) but did not correlate with stemness-associated markers. Further analyses in CRC cells lines as well as xenograft mice including forced overexpression of OLFM4 revealed that OLFM4 neither altered the expression of markers of stemness nor epithelial-mesenchymal transition, nor did OLFM4 itself drive proliferation, migration, or colony formation, which are all prerequisites of carcinogenesis and tumor progression. In line with this, we found no significant correlation between OLFM4 expression, metastasis, and patient survival. In summary, expression of OLFM4 in human CRC seems to be characteristic of differentiation marker expression in CRC but is not a driver of carcinogenesis nor metastatic spread.
Collapse
Affiliation(s)
- Stefanie Jaitner
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Elise Pretzsch
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-Universität München, Munich, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Munich, Heidelberg, Germany
| | - Jens Neumann
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Munich, Heidelberg, Germany
| | - Achim Schäffauer
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Matthias Schiemann
- Institute of Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany.,Clinical Cooperation Group Immune Monitoring, Helmholtz Center Munich (Neuherberg) and Technische Universität München, Munich, Germany
| | - Martin Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jörg Kumbrink
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Munich, Heidelberg, Germany
| | - Sarah Schwitalla
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Florian R Greten
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Munich, Heidelberg, Germany.,Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Lydia Brandl
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Frederick Klauschen
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Munich, Heidelberg, Germany
| | - David Horst
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Kirchner
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Munich, Heidelberg, Germany
| | - Andreas Jung
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Munich, Heidelberg, Germany
| |
Collapse
|
8
|
Xu C, Hu X, Fan Y, Zhang L, Gao Z, Cai C. Wif1 Mediates Coordination of Bone Morphogenetic Protein and Wnt Signaling in Neural and Glioma Stem Cells. Cell Transplant 2022; 31:9636897221134540. [PMID: 36324293 PMCID: PMC9634200 DOI: 10.1177/09636897221134540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Wnts, bone morphogenetic protein (BMP), and fibroblast growth factor (FGF) are
paracrine signaling pathways implicated in the niche control of stem cell fate
decisions. BMP-on and Wnt-off are the dominant quiescent niche signaling
pathways in many cell types, including neural stem cells (NSCs). However, among
the multiple inhibitory family members of the Wnt pathway, those with direct
action after BMP4 stimulation in NSCs remain unclear. We examined 11 Wnt
inhibitors in NSCs after BMP4 treatment. Wnt inhibitory factor 1 (Wif1) has been
identified as the main factor reacting to BMP4 stimuli. RNA sequencing confirmed
that Wif1 was markedly upregulated after BMP4 treatment in different gene
expression analyses. Similar to the functional role of BMP4, Wif1 significantly
decreased the cell cycle of NSCs and significantly inhibited cell proliferation
(P < 0.05). Combined treatment with BMP4 and Wif1
significantly enhanced the inhibition of cell growth compared with the single
treatment (P < 0.05). Wif1 expression was clearly lower in
glioblastoma and low-grade glioma samples than in normal samples
(P < 0.05). A functional analysis revealed that both
BMP4 and Wif1 could decrease glioma cell growth. These effects were abrogated by
the BMP inhibitor Noggin. The collective findings demonstrate that Wif1 plays a
key role in quiescent NSC homeostasis and glioma cell growth downstream of
BMP-on signaling. The functional roles of Wif1/BMP4 in glioma cells may provide
a technical basis for regenerative medicine, drug discovery, and personal
molecular therapy in future clinical treatments.
Collapse
Affiliation(s)
- Congdi Xu
- Fundamental Research Center, Shanghai
YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of
Medicine, Tongji University, Shanghai, China
| | - Xinyu Hu
- Fundamental Research Center, Shanghai
YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of
Medicine, Tongji University, Shanghai, China,Institute for Molecules and Materials,
Radboud University, Nijmegen, The Netherlands
| | - Yantao Fan
- Fundamental Research Center, Shanghai
YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of
Medicine, Tongji University, Shanghai, China,Institute of Geriatrics (Shanghai
University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s
Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
| | - Ling Zhang
- The First Rehabilitation Hospital of
Shanghai, School of Medicine, Tongji University, Shanghai, China
| | - Zhengliang Gao
- Fundamental Research Center, Shanghai
YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of
Medicine, Tongji University, Shanghai, China,Institute of Geriatrics (Shanghai
University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s
Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
| | - Chunhui Cai
- Fundamental Research Center, Shanghai
YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of
Medicine, Tongji University, Shanghai, China,Institute of Geriatrics (Shanghai
University), Affiliated Nantong Hospital of Shanghai University (The Sixth People’s
Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China,Chunhui Cai, Fundamental Research Center,
Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation
Center), School of Medicine, Tongji University, Shanghai 200001, China.
| |
Collapse
|
9
|
Ben Amar D, Thoinet K, Villalard B, Imbaud O, Costechareyre C, Jarrosson L, Reynaud F, Novion Ducassou J, Couté Y, Brunet JF, Combaret V, Corradini N, Delloye-Bourgeois C, Castellani V. Environmental cues from neural crest derivatives act as metastatic triggers in an embryonic neuroblastoma model. Nat Commun 2022; 13:2549. [PMID: 35538114 PMCID: PMC9091272 DOI: 10.1038/s41467-022-30237-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 04/21/2022] [Indexed: 12/04/2022] Open
Abstract
Embryonic malignant transformation is concomitant to organogenesis, often affecting multipotent and migratory progenitors. While lineage relationships between malignant cells and their physiological counterparts are extensively investigated, the contribution of exogenous embryonic signals is not fully known. Neuroblastoma (NB) is a childhood malignancy of the peripheral nervous system arising from the embryonic trunk neural crest (NC) and characterized by heterogeneous and interconvertible tumor cell identities. Here, using experimental models mimicking the embryonic context coupled to proteomic and transcriptomic analyses, we show that signals released by embryonic sympathetic ganglia, including Olfactomedin-1, induce NB cells to shift from a noradrenergic to mesenchymal identity, and to activate a gene program promoting NB metastatic onset and dissemination. From this gene program, we extract a core signature specifically shared by metastatic cancers with NC origin. This reveals non-cell autonomous embryonic contributions regulating the plasticity of NB identities and setting pro-dissemination gene programs common to NC-derived cancers.
Collapse
Affiliation(s)
- Dounia Ben Amar
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller
| | - Karine Thoinet
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller
| | - Benjamin Villalard
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller
| | - Olivier Imbaud
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller
| | | | | | - Florie Reynaud
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller
| | - Julia Novion Ducassou
- University Grenoble Alpes, INSERM, CEA, UMR BioSanté U1292, CNRS, CEA, FR2048 38000, Grenoble, France
| | - Yohann Couté
- University Grenoble Alpes, INSERM, CEA, UMR BioSanté U1292, CNRS, CEA, FR2048 38000, Grenoble, France
| | - Jean-François Brunet
- Institut de Biologie de l'ENS (IBENS), Inserm, CNRS, École normale supérieure, PSL Research University, Paris, France
| | - Valérie Combaret
- Laboratory of Translational Research, Léon Bérard Centre, Lyon, France
| | - Nadège Corradini
- Departments of Oncology and Clinical Research, Centre Léon Berard and Institut d'Hématologie et d'Oncologie Pédiatrique, Lyon, France
| | - Céline Delloye-Bourgeois
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller.
| | - Valérie Castellani
- University of Lyon, University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, NeuroMyoGene Institute, 69008, Lyon, France, 8 avenue Rockefeller.
| |
Collapse
|
10
|
Shimoide T, Kawao N, Morita H, Ishida M, Takafuji Y, Kaji H. Roles of Olfactomedin 1 in Muscle and Bone Alterations Induced by Gravity Change in Mice. Calcif Tissue Int 2020; 107:180-190. [PMID: 32462291 DOI: 10.1007/s00223-020-00710-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/19/2020] [Indexed: 02/08/2023]
Abstract
Microgravity causes both muscle and bone loss. Although we previously revealed that gravity change influences muscle and bone through the vestibular system in mice, its detailed mechanism has not been elucidated. In this study, we investigated the roles of olfactomedin 1 (OLFM1), whose expression was upregulated during hypergravity in the soleus muscle, in mouse bone cells. Vestibular lesion significantly blunted OLFM1 expression in the soleus muscle and serum OLFM1 levels enhanced by hypergravity in mice. Moreover, a phosphatidylinositol 3-kinase inhibitor antagonized shear stress-enhanced OLFM1 expression in C2C12 myotubes. As for the effects of OLFM1 on bone cells, OLFM1 inhibited osteoclast formation from mouse bone marrow cells and mouse preosteoclastic RAW264.7 cells. Moreover, OLFM1 suppressed RANKL expression and nuclear factor-κB signaling in mouse osteoblasts. Serum OLFM1 levels were positively related to OLFM1 mRNA levels in the soleus muscle and trabecular bone mineral density of mice. In conclusion, we first showed that OLFM1 suppresses osteoclast formation and RANKL expression in mouse cells.
Collapse
Affiliation(s)
- Takeshi Shimoide
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osakasayama, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, Japan
| | - Naoyuki Kawao
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osakasayama, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, Japan
| | - Hironobu Morita
- Department of Physiology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | - Masayoshi Ishida
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osakasayama, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, Japan
| | - Yoshimasa Takafuji
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osakasayama, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, Japan
| | - Hiroshi Kaji
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osakasayama, 377-2 Ohnohigashi, Osakasayama, Osaka, 589-8511, Japan.
| |
Collapse
|
11
|
Carlo SE, Martinez-Baladejo MT, Santiago-Cornier A, Arciniegas-Medina N. 9q34 & 16p13 chromosome duplications in autism. AME Case Rep 2020; 4:17. [PMID: 32793859 DOI: 10.21037/acr.2020.03.07] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 02/28/2020] [Indexed: 11/06/2022]
Abstract
Epigenetic mechanisms, genetic factors, and environment influence the diversity of phenotypes developed in various diseases. Duplications in several chromosomes are well characterized in the scientific literature, but partial duplications, in some cases, present with milder forms of a disease and are yet to be understood. Fortunately, the identification of genetic diseases has now become more feasible due to several cytogenetic techniques such as microarray analysis and karyotyping. With these tools, together with other laboratory results and clinical examination, we are able to report the first case in the medical literature of double partial trisomy of chromosome 9q34 and 16p13.
Collapse
Affiliation(s)
- Simon E Carlo
- Department of Biochemistry, Ponce Health Sciences University, Ponce.,Department of Medicine, Ponce Health Sciences University, Ponce.,SER de Puerto Rico, Ponce.,Mayagüez Medical Center, Mayaguez, Ponce
| | | | | | | |
Collapse
|
12
|
Pronker MF, van den Hoek H, Janssen BJC. Design and structural characterisation of olfactomedin-1 variants as tools for functional studies. BMC Mol Cell Biol 2019; 20:50. [PMID: 31726976 PMCID: PMC6857237 DOI: 10.1186/s12860-019-0232-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/10/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Olfactomedin-1 (Olfm1; also known as Noelin or Pancortin) is a highly-expressed secreted brain and retina protein and its four isoforms have different roles in nervous system development and function. Structural studies showed that the long Olfm1 isoform BMZ forms a disulfide-linked tetramer with a V-shaped architecture. The tips of the Olfm1 "V" each consist of two C-terminal β-propeller domains that enclose a calcium binding site. Functional characterisation of Olfm1 may be aided by new biochemical tools derived from these core structural elements. RESULTS Here we present the production, purification and structural analysis of three novel monomeric, dimeric and tetrameric forms of mammalian Olfm1 for functional studies. We characterise these constructs structurally by high-resolution X-ray crystallography and small-angle X-ray scattering. The crystal structure of the Olfm1 β-propeller domain (to 1.25 Å) represents the highest-resolution structure of an olfactomedin family member to date, revealing features such as a hydrophilic tunnel containing water molecules running into the core of the domain where the calcium binding site resides. The shorter Olfactomedin-1 isoform BMY is a disulfide-linked tetramer with a shape similar to the corresponding region in the longer BMZ isoform. CONCLUSIONS These recombinantly-expressed protein tools should assist future studies, for example of biophysical, electrophysiological or morphological nature, to help elucidate the functions of Olfm1 in the mature mammalian brain. The control over the oligomeric state of Olfm1 provides a firm basis to better understand the role of Olfm1 in the (trans-synaptic) tethering or avidity-mediated clustering of synaptic receptors such as post-synaptic AMPA receptors and pre-synaptic amyloid precursor protein. In addition, the variation in domain composition of these protein tools provides a means to dissect the Olfm1 regions important for receptor binding.
Collapse
Affiliation(s)
- Matti F Pronker
- MRC Laboratory of Molecular Biology, Division of Neurobiology, Francis Crick Avenue, Cambridge, CB2 0QH, UK. .,Bijvoet Center for Biomolecular Research, Utrecht University, Crystal and Structural Chemistry, Kruytgebouw, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - Hugo van den Hoek
- Bijvoet Center for Biomolecular Research, Utrecht University, Crystal and Structural Chemistry, Kruytgebouw, Padualaan 8, 3584 CH, Utrecht, The Netherlands.,Department of Molecular Structural Biology, Max Planck institute for Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany
| | - Bert J C Janssen
- Bijvoet Center for Biomolecular Research, Utrecht University, Crystal and Structural Chemistry, Kruytgebouw, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| |
Collapse
|
13
|
Garza-Rodríguez ML, González-Álvarez R, Mendoza Alfaro RE, Pérez-Ibave DC, Perez-Maya AA, Luna-Muñoz M, Mohamed-Noriega K, Arámburo-De-La-Hoz C, Aguilera González CJ, Rodriguez Sanchez IP. Olfactomedin-like 2 A and B (OLFML2A and OLFML2B) profile expression in the retina of spotted gar (Lepisosteus oculatus) and bioinformatics mining. FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:1575-1587. [PMID: 31111317 DOI: 10.1007/s10695-019-00647-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/23/2019] [Indexed: 06/09/2023]
Abstract
Olfactomedin-like (OLFML) proteins are members of the olfactomedin domain-containing secreted glycoprotein (OLF) family. OLFML2A and OLFML2B are representative molecules of these glycoproteins. Olfactomedins are critical for the development and functional organization of the nervous system and retina, which is a highly conserved structure in vertebrates, having almost identical anatomical and physiological characteristics in multiple taxa. Spotted gar, a member of the Lepisosteidae family, is a freshwater fish that inhabits rivers, bayous, swamps, and brackish waters. Recently, the complete genome has been sequenced, providing a unique bridge between fish medical models to human biology, making it an excellent animal model. This study was aimed to understanding the evolution OLFML2A and OLFML2B in the retina of spotted gar through looking for the expression of these genes. Spotted gar retina was analyzed with hematoxylin-eosin staining assays to provide an overall view of the retina structure and an immunofluorescence assay to identify OLFML2A and OLFML2B protein expression. A phylogenetic tree was created using the neighbor-joining method. Forces that direct the evolution of the fish genes were tested. Spotted gar retina, as in other vertebrates, is made of several layers. OLFML2A and OLFML2B proteins were detected in the rod and cone photoreceptor layer (PRL), outer nuclear layer (ONL), and inner nuclear layer (INL). Phylogenetic tree analysis confirms the orthology within the OLFML2A gene. Purifying selection is the evolutionary force that directs the OLFML2A genes. OLFML2A genes have a well-conserved function over time and species.
Collapse
Affiliation(s)
- María Lourdes Garza-Rodríguez
- Universidad Autónoma de Nuevo León, Hospital Universitario "Dr. José Eleuterio González," Servicio de Oncología, Monterrey, Nuevo León, Mexico
| | | | - Roberto Eduardo Mendoza Alfaro
- Facultad de Ciencias Biológicas, Departamento de Ecología, Laboratorio de Ecofisiología, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| | - Diana Cristina Pérez-Ibave
- Universidad Autónoma de Nuevo León, Hospital Universitario "Dr. José Eleuterio González," Servicio de Oncología, Monterrey, Nuevo León, Mexico
| | - Antonio Ali Perez-Maya
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Bioquímica y Medicina Molecular, Monterrey, Nuevo León, Mexico
| | - Maricela Luna-Muñoz
- Instituo de Neurobiología, Departamento de Neurobiología Celular y Molecular, Universidad Nacional Autónoma de México, Juriquilla, Queretaro, Mexico
| | - Karim Mohamed-Noriega
- Departamento de Oftalmología, Universidad Autónoma de Nuevo León, Hospital Universitario "Dr. José Eleuterio González", Monterrey, Nuevo León, Mexico
| | - Carlos Arámburo-De-La-Hoz
- Instituo de Neurobiología, Departamento de Neurobiología Celular y Molecular, Universidad Nacional Autónoma de México, Juriquilla, Queretaro, Mexico
| | - Carlos Javier Aguilera González
- Facultad de Ciencias Biológicas, Departamento de Ecología, Laboratorio de Ecofisiología, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| | - Iram Pablo Rodriguez Sanchez
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Laboratorio de Fisiología Molecular y Estructural, Ave. Pedro de Alba s/n cruz con Ave. Manuel L. Barragán, 66455, San Nicolás de los Garza, Nuevo León, México.
| |
Collapse
|
14
|
Bissen D, Foss F, Acker-Palmer A. AMPA receptors and their minions: auxiliary proteins in AMPA receptor trafficking. Cell Mol Life Sci 2019; 76:2133-2169. [PMID: 30937469 PMCID: PMC6502786 DOI: 10.1007/s00018-019-03068-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/12/2019] [Accepted: 03/07/2019] [Indexed: 12/12/2022]
Abstract
To correctly transfer information, neuronal networks need to continuously adjust their synaptic strength to extrinsic stimuli. This ability, termed synaptic plasticity, is at the heart of their function and is, thus, tightly regulated. In glutamatergic neurons, synaptic strength is controlled by the number and function of AMPA receptors at the postsynapse, which mediate most of the fast excitatory transmission in the central nervous system. Their trafficking to, at, and from the synapse, is, therefore, a key mechanism underlying synaptic plasticity. Intensive research over the last 20 years has revealed the increasing importance of interacting proteins, which accompany AMPA receptors throughout their lifetime and help to refine the temporal and spatial modulation of their trafficking and function. In this review, we discuss the current knowledge about the roles of key partners in regulating AMPA receptor trafficking and focus especially on the movement between the intracellular, extrasynaptic, and synaptic pools. We examine their involvement not only in basal synaptic function, but also in Hebbian and homeostatic plasticity. Included in our review are well-established AMPA receptor interactants such as GRIP1 and PICK1, the classical auxiliary subunits TARP and CNIH, and the newest additions to AMPA receptor native complexes.
Collapse
Affiliation(s)
- Diane Bissen
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
- Max Planck Institute for Brain Research, Max von Laue Str. 4, 60438, Frankfurt am Main, Germany
| | - Franziska Foss
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany.
- Max Planck Institute for Brain Research, Max von Laue Str. 4, 60438, Frankfurt am Main, Germany.
- Cardio-Pulmonary Institute (CPI), Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
15
|
Poggi L, Casarosa S, Carl M. An Eye on the Wnt Inhibitory Factor Wif1. Front Cell Dev Biol 2018; 6:167. [PMID: 30574494 PMCID: PMC6292148 DOI: 10.3389/fcell.2018.00167] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022] Open
Abstract
The coordinated interplay between extrinsic activating and repressing cell signaling molecules is pivotal for embryonic development and subsequent tissue homeostasis. This is well exemplified by studies on the evolutionarily conserved Wnt signaling pathways. Tight temporal and spatial regulation of Wnt signaling activity is required throughout lifetime, from maternal stages before gastrulation until and throughout adulthood. Outside cells, the action of numerous Wnt ligands is counteracted and fine-tuned by only a handful of well characterized secreted inhibitors, such as for instance Dickkopf, secreted Frizzled Related Proteins and Cerberus. Here, we give an overview of our current understanding of another secreted Wnt signaling antagonist, the Wnt inhibitory factor Wif1. Wif1 can directly interact with various Wnt ligands and inhibits their binding to membrane bound receptors. Epigenetic promoter methylation of Wif1, leading to silencing of its transcription and concomitant up-regulation of Wnt signaling, is a common feature during cancer progression. Furthermore, an increasing number of reports describe Wif1 involvement in regulating processes during embryonic development, which so far has not received as much attention. We will summarize our knowledge on Wif1 function and its mode of action with a particular focus on the zebrafish (Danio rerio). In addition, we highlight the potential of Wif1 research to understand and possibly influence mechanisms underlying eye diseases and regeneration.
Collapse
Affiliation(s)
- Lucia Poggi
- Laboratory of Molecular and Cellular Ophthalmology, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Simona Casarosa
- Laboratory of Neural Development and Regeneration, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Matthias Carl
- Laboratory of Translational Neurogenetics, Centre for Integrative Biology, University of Trento, Trento, Italy
| |
Collapse
|
16
|
Neidert N, von Ehr A, Zöller T, Spittau B. Microglia-Specific Expression of Olfml3 Is Directly Regulated by Transforming Growth Factor β1-Induced Smad2 Signaling. Front Immunol 2018; 9:1728. [PMID: 30093905 PMCID: PMC6070609 DOI: 10.3389/fimmu.2018.01728] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 07/12/2018] [Indexed: 11/13/2022] Open
Abstract
Microglia maturation takes place during the postnatal weeks and is characterized by the establishment of a unique microglia-specific gene expression pattern. Tmem119, Fcrls, Hexb, and Olfml3 have been identified among these microglia-specific genes. Transforming growth factor β1 (TGFβ1) has been reported as a critical factor for microglia maturation and maintenance and active TGFβ signaling precedes the inductions of microglial gene expression. In this study, we demonstrate Olfml3 expression in adult microglia and further provide evidence that TGFβ1 induces upregulation of Olfml3 expression in postnatal microglia. Using chromatin immunoprecipitation and microglia-specific silencing of TGFβ signaling in vitro and in vivo, we in clearly show that Olfml3 is a direct TGFβ1/Smad2 target gene. Together, our data underline the importance of TGFβ1 as a critical regulator of microglia functions and microglia maturation and further broaden our understanding of TGFβ1-mediated effects on the resident immune cells of the central nervous system.
Collapse
Affiliation(s)
- Nicolas Neidert
- Department of Molecular Embryology, Faculty of Medicine, Institute for Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | - Alexander von Ehr
- Department of Molecular Embryology, Faculty of Medicine, Institute for Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | - Tanja Zöller
- Department of Molecular Embryology, Faculty of Medicine, Institute for Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | - Björn Spittau
- Department of Molecular Embryology, Faculty of Medicine, Institute for Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany.,Institute of Anatomy, University of Rostock, Rostock, Germany
| |
Collapse
|
17
|
Nakaya N, Sultana A, Tomarev SI. Impaired AMPA receptor trafficking by a double knockout of zebrafish olfactomedin1a/b. J Neurochem 2017; 143:635-644. [PMID: 28975619 DOI: 10.1111/jnc.14231] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/15/2017] [Accepted: 09/27/2017] [Indexed: 01/06/2023]
Abstract
The olfm1a and olfm1b genes in zebrafish encode conserved secreted glycoproteins. These genes are preferentially expressed in the brain and retina starting from 16 h post-fertilization until adulthood. Functions of the Olfm1 gene is still unclear. Here, we produced and analyzed a null zebrafish mutant of both olfm1a and olfm1b genes (olfm1 null). olfm1 null fish were born at a normal Mendelian ratio and showed normal body shape and fertility as well as no visible defects from larval stages to adult. Olfm1 proteins were preferentially localized in the synaptosomes of the adult brain. Olfm1 co-immunoprecipitated with GluR2 and soluble NSF attachment protein receptor complexes indicating participation of Olfm1 in both pre- and post-synaptic events. Phosphorylation of GluR2 was not changed while palmitoylation of GluR2 was decreased in the brain synaptosomal membrane fraction of olfm1 null compared with wt fish. The levels of GluR2, SNAP25, flotillin1, and VAMP2 were markedly reduced in the synaptic microdomain of olfm1 null brain compared with wt. The internalization of GluR2 in retinal cells and the localization of VAMP2 in brain synaptosome were modified by olfm1 null mutation. This indicates that Olfm1 may regulate receptor trafficking from the intracellular compartments to the synaptic membrane microdomain, partly through the alteration of post-translational GluR2 modifications such as palmitoylation. Olfm1 may be considered a novel regulator of the composition and function of the α-amino-3-hydroxy-5-methylisoxazole-4-propionate receptor complex.
Collapse
Affiliation(s)
- Naoki Nakaya
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, NIH, Bethesda, Maryland, USA
| | - Afia Sultana
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, NIH, Bethesda, Maryland, USA
| | - Stanislav I Tomarev
- Section of Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
18
|
Binelli A, Del Giacco L, Santo N, Bini L, Magni S, Parolini M, Madaschi L, Ghilardi A, Maggioni D, Ascagni M, Armini A, Prosperi L, Landi C, La Porta C, Della Torre C. Carbon nanopowder acts as a Trojan-horse for benzo(α)pyrene in Danio rerio embryos. Nanotoxicology 2017; 11:371-381. [DOI: 10.1080/17435390.2017.1306130] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- A. Binelli
- Department of Biosciences, University of Milan, Milan, Italy
| | - L. Del Giacco
- Department of Biosciences, University of Milan, Milan, Italy
| | - N. Santo
- Department of Biosciences, University of Milan, Milan, Italy
| | - L. Bini
- Department of Life Science, University of Siena, Siena, Italy
| | - S. Magni
- Department of Biosciences, University of Milan, Milan, Italy
| | - M. Parolini
- Department of Biosciences, University of Milan, Milan, Italy
| | - L. Madaschi
- Department of Biosciences, University of Milan, Milan, Italy
| | - A. Ghilardi
- Department of Biosciences, University of Milan, Milan, Italy
| | - D. Maggioni
- Department of Chemistry, University of Milan, Milan, Italy
| | - M. Ascagni
- Department of Biosciences, University of Milan, Milan, Italy
| | - A. Armini
- Department of Life Science, University of Siena, Siena, Italy
| | - L. Prosperi
- Department of Biosciences, University of Milan, Milan, Italy
| | - C. Landi
- Department of Life Science, University of Siena, Siena, Italy
| | - C. La Porta
- Department of Biosciences, University of Milan, Milan, Italy
| | - C. Della Torre
- Department of Biosciences, University of Milan, Milan, Italy
| |
Collapse
|
19
|
Pérez-Ibave DC, González-Alvarez R, de La Luz Martinez-Fierro M, Ruiz-Ayma G, Luna-Muñoz M, Martínez-De-Villarreal LE, De Lourdes Garza-Rodríguez M, Reséndez-Pérez D, Mohamed-Noriega J, Garza-Guajardo R, Bautista-De-Lucío VM, Mohamed-Noriega K, Barboza-Quintana O, Arámburo-De-La-Hoz C, Barrera-Saldaña HA, Rodríguez-Sánchez IP. Olfactomedin-like 2 A and B (OLFML2A and OLFML2B) expression profile in primates (human and baboon). Biol Res 2016; 49:44. [PMID: 27821182 PMCID: PMC5100274 DOI: 10.1186/s40659-016-0101-8] [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: 04/27/2016] [Accepted: 09/24/2016] [Indexed: 12/12/2022] Open
Abstract
Background The olfactomedin-like domain (OLFML) is present in at least four families of proteins, including OLFML2A and OLFML2B, which are expressed in adult rat retina cells. However, no expression of their orthologous has ever been reported in human and baboon. Objective The aim of this study was to investigate the expression of OLFML2A and OLFML2B in ocular tissues of baboons (Papio hamadryas) and humans, as a key to elucidate OLFML function in eye physiology. Methods OLFML2A and OLFML2B cDNA detection in ocular tissues of these species was performed by RT-PCR. The amplicons were cloned and sequenced, phylogenetically analyzed and their proteins products were confirmed by immunofluorescence assays. Results OLFML2A and OLFML2B transcripts were found in human cornea, lens and retina and in baboon cornea, lens, iris and retina. The baboon OLFML2A and OLFML2B ORF sequences have 96% similarity with their human’s orthologous. OLFML2A and OLFML2B evolution fits the hypothesis of purifying selection. Phylogenetic analysis shows clear orthology in OLFML2A genes, while OLFML2B orthology is not clear. Conclusions Expression of OLFML2A and OLFML2B in human and baboon ocular tissues, including their high similarity, make the baboon a powerful model to deduce the physiological and/or metabolic function of these proteins in the eye.
Collapse
Affiliation(s)
- Diana Cristina Pérez-Ibave
- Servicio de Oncología, Universidad Autónoma de Nuevo León, Hospital Universitario "Dr. José Eleuterio González", Monterrey, Nuevo León, Mexico
| | | | - Margarita de La Luz Martinez-Fierro
- Unidad Académica de Medicina Humana y Ciencias de la Salud, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km.6. Ejido La Escondida, Zacatecas, Mexico
| | - Gabriel Ruiz-Ayma
- Departamento de Ecología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, 66451, San Nicolás de los Garza, Nuevo León, Mexico
| | - Maricela Luna-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, Mexico
| | - Laura Elia Martínez-De-Villarreal
- Departamento de Genética, Universidad Autónoma de Nuevo León, Hospital Universitario "Dr. José Eleuterio González", 64460, Monterrey, Nuevo León, Mexico
| | - María De Lourdes Garza-Rodríguez
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Diana Reséndez-Pérez
- Departamento de Biología Celular y Genética, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Jibran Mohamed-Noriega
- Departamento de Oftalmología, Universidad Autónoma de Nuevo León Hospital Universitario "Dr. José Eleuterio González", Monterrey, Nuevo León, Mexico
| | - Raquel Garza-Guajardo
- Servicio de Anatomía Patológica y Citopatología, Universidad Autónoma de Nuevo León, Hospital Universitario "Dr. José Eleuterio González", Monterrey, Nuevo León, Mexico
| | - Víctor Manuel Bautista-De-Lucío
- Departamento de Bioquímica y Medicina Molecular, Instituto de Oftalmología. Fundación de Asistencia Privada Conde de Valenciana IAP, Mexico, Mexico
| | - Karim Mohamed-Noriega
- Departamento de Oftalmología, Universidad Autónoma de Nuevo León Hospital Universitario "Dr. José Eleuterio González", Monterrey, Nuevo León, Mexico
| | - Oralia Barboza-Quintana
- Servicio de Anatomía Patológica y Citopatología, Universidad Autónoma de Nuevo León, Hospital Universitario "Dr. José Eleuterio González", Monterrey, Nuevo León, Mexico
| | - Carlos Arámburo-De-La-Hoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, Mexico
| | - Hugo Alberto Barrera-Saldaña
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Irám Pablo Rodríguez-Sánchez
- Departamento de Genética, Universidad Autónoma de Nuevo León, Hospital Universitario "Dr. José Eleuterio González", 64460, Monterrey, Nuevo León, Mexico.
| |
Collapse
|
20
|
Mutated olfactomedin 1 in the interphotoreceptor matrix of the mouse retina causes functional deficits and vulnerability to light damage. Histochem Cell Biol 2016; 147:453-469. [PMID: 27787612 DOI: 10.1007/s00418-016-1510-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2016] [Indexed: 12/24/2022]
Abstract
Olfactomedin 1 (OLFM1) is a secreted glycoprotein and member of the olfactomedin protein family, which is preferentially expressed in various areas throughout the central nervous system. To learn about the functional properties of OLFM1 in the eye, we investigated its localization in the mouse and pig eye. In addition, we analyzed the ocular phenotype of Olfm1 mutant mice in which 52 amino acids were deleted in the central part (M2 region) of OLFM1. OLFM1 was detected in cornea, sclera, retina, and optic nerve of both wild-type and Olfm1 mutant littermates. By immunohistochemistry and double labeling with the lectin peanut agglutinin, OLFM1 was found in the interphotoreceptor matrix (IPM) of mouse and pig retina where it was directly localized to the inner segments of photoreceptors. Western blotting confirmed the presence of the OLFM1 isoforms pancortin 1 (BMY) and pancortin 2 (BMZ) in the IPM. The retinal phenotype of Olfm1 mutant mice did not obviously differ from that of wild-type littermates. In addition, outer nuclear layer (ONL) and total retinal thickness were not different, and the same was true for the area of the optic nerve in cross sections. Functional changes were observed though by electroretinography, which showed significantly lower a- and b-wave amplitudes in Olfm1 mutant mice when compared to age-matched wild-type mice. When light damage experiments were performed as an experimental paradigm of photoreceptor apoptosis, significantly more TUNEL-positive cells were observed in Olfm1 mutant mice 30 h after light exposure. One week after light exposure, the ONL was significantly thinner in Olfm1 mutant mice than in wild-type littermates indicating increased photoreceptor loss. No differences were observed when rhodopsin turnover or ERK1/2 signaling was investigated. We conclude that OLFM1 is a newly identified IPM molecule that serves an important role for photoreceptor homeostasis, which is significantly compromised in the eyes of Olfm1 mutant mice.
Collapse
|
21
|
Olfactomedin 4 deletion induces colon adenocarcinoma in Apc Min/+ mice. Oncogene 2016; 35:5237-5247. [PMID: 26973250 PMCID: PMC5057043 DOI: 10.1038/onc.2016.58] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 01/27/2016] [Accepted: 02/02/2016] [Indexed: 12/13/2022]
Abstract
Colon carcinogenesis is a multiple-step process involving the accumulation of a series of genetic and epigenetic alterations. The most commonly initiating event of intestinal carcinogenesis is mutation of the adenomatous polyposis coli (APC) gene, which leads to activation of the Wnt/β-catenin pathway. Olfactomedin 4 (OLFM4) has emerged as an intestinal stem-cell marker, but its biological function in the intestine remains to be determined. Here we show that Olfm4 deletion induced colon adenocarcinoma in the distal colon of ApcMin/+ mice. Mechanistically, we found that OLFM4 is a target gene of the Wnt/β-catenin pathway and can downregulate β-catenin signaling by competing with Wnt ligands for binding to Frizzled receptors, as well as by inhibition of the Akt-GSK-3β (Akt-glycogen synthase kinase-3β) pathway. We have shown that both Wnt and nuclear factor-κB (NF-κB) signaling were boosted in tumor tissues of Apc Olfm4 double-mutant mice. These data establish OLFM4 as a critical negative regulator of the Wnt/β-catenin and NF-κB pathways that inhibits colon-cancer development initiated by APC mutation. In addition, Olfm4 deletion significantly enhanced intestinal-crypt proliferation and inflammation induced by azoxymethane/dextran sodium sulfate. Thus, OLFM4 has an important role in the regulation of intestinal inflammation and tumorigenesis, and could be a potential therapeutic target for intestinal malignant tumors. Unlike the human colonic epithelium, the mouse colonic epithelium does not express OLFM4, but nevertheless, systemic OLFM4 deletion promotes colon tumorigenesis and that loss from mucosal neutrophils may have a role to play.
Collapse
|
22
|
So KH, Kodithuwakku SP, Kottawatta KS, Li RH, Chiu PC, Cheung AN, Ng EH, Yeung WS, Lee KF. Human chorionic gonadotropin stimulates spheroid attachment on fallopian tube epithelial cells through the mitogen-activated protein kinase pathway and down-regulation of olfactomedin-1. Fertil Steril 2015; 104:474-82. [DOI: 10.1016/j.fertnstert.2015.04.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 04/21/2015] [Accepted: 04/21/2015] [Indexed: 12/23/2022]
|
23
|
Hill SE, Donegan RK, Nguyen E, Desai TM, Lieberman RL. Molecular Details of Olfactomedin Domains Provide Pathway to Structure-Function Studies. PLoS One 2015; 10:e0130888. [PMID: 26121352 PMCID: PMC4488277 DOI: 10.1371/journal.pone.0130888] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 05/26/2015] [Indexed: 11/18/2022] Open
Abstract
Olfactomedin (OLF) domains are found within extracellular, multidomain proteins in numerous tissues of multicellular organisms. Even though these proteins have been implicated in human disorders ranging from cancers to attention deficit disorder to glaucoma, little is known about their structure(s) and function(s). Here we biophysically, biochemically, and structurally characterize OLF domains from H. sapiens olfactomedin-1 (npoh-OLF, also called noelin, pancortin, OLFM1, and hOlfA), and M. musculus gliomedin (glio-OLF, also called collomin, collmin, and CRG-L2), and compare them with available structures of myocilin (myoc-OLF) recently reported by us and R. norvegicus glio-OLF and M. musculus latrophilin-3 (lat3-OLF) by others. Although the five-bladed β-propeller architecture remains unchanged, numerous physicochemical characteristics differ among these OLF domains. First, npoh-OLF and glio-OLF exhibit prominent, yet distinct, positive surface charges and copurify with polynucleotides. Second, whereas npoh-OLF and myoc-OLF exhibit thermal stabilities typical of human proteins near 55°C, and most myoc-OLF variants are destabilized and highly prone to aggregation, glio-OLF is nearly 20°C more stable and significantly more resistant to chemical denaturation. Phylogenetically, glio-OLF is most similar to primitive OLFs, and structurally, glio-OLF is missing distinguishing features seen in OLFs such as the disulfide bond formed by N- and C- terminal cysteines, the sequestered Ca2+ ion within the propeller central hydrophilic cavity, and a key loop-stabilizing cation-π interaction on the top face of npoh-OLF and myoc-OLF. While deciphering the explicit biological functions, ligands, and binding partners for OLF domains will likely continue to be a challenging long-term experimental pursuit, we used structural insights gained here to generate a new antibody selective for myoc-OLF over npoh-OLF and glio-OLF as a first step in overcoming the impasse in detailed functional characterization of these biomedically important protein domains.
Collapse
Affiliation(s)
- Shannon E. Hill
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Rebecca K. Donegan
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Elaine Nguyen
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Tanay M. Desai
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Raquel L. Lieberman
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
24
|
Yang YHC, Wills QF, Johnson JD. A live-cell, high-content imaging survey of 206 endogenous factors across five stress conditions reveals context-dependent survival effects in mouse primary beta cells. Diabetologia 2015; 58:1239-49. [PMID: 25773404 PMCID: PMC4415993 DOI: 10.1007/s00125-015-3552-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/10/2015] [Indexed: 12/22/2022]
Abstract
AIMS/HYPOTHESIS Beta cell death is a hallmark of diabetes. It is not known whether specific cellular stresses associated with type 1 or type 2 diabetes require specific factors to protect pancreatic beta cells. No systematic comparison of endogenous soluble factors in the context of multiple pro-apoptotic conditions has been published. METHODS Primary mouse islet cells were cultured in conditions mimicking five type 1 or type 2 diabetes-related stresses: basal 5 mmol/l glucose, cytokine cocktail (25 ng/ml TNF-α, 10 ng/ml IL-1β, 10 ng/ml IFN-γ), 1 μmol/l thapsigargin, 1.5 mmol/l palmitate and 20 mmol/l glucose (all in the absence of serum). We surveyed the effects of a library of 206 endogenous factors (selected based on islet expression of their receptors) on islet cell survival through multi-parameter, live-cell imaging. RESULTS Our survey pointed to survival factors exhibiting generalised protective effects across conditions meant to model different types of diabetes and stages of the diseases. For example, our survey and follow-up experiments suggested that OLFM1 is a novel protective factor for mouse and human beta cells across multiple conditions. Most strikingly, we also found specific protective survival factors for each model stress condition. For example, semaphorin4A (SEMA4A) was toxic to islet cells in the serum-free baseline and serum-free 20 mmol/l glucose conditions, but protective in the context of lipotoxicity. Rank product testing supported the consistency of our observations. CONCLUSIONS/INTERPRETATION Collectively, our survey reveals previously unidentified islet cell survival factors and suggest their potential utility in individualised medicine.
Collapse
Affiliation(s)
- Yu Hsuan Carol Yang
- Department of Cellular and Physiological Sciences, Faculty of Medicine, Diabetes Research Group, Life Sciences Institute, University of British Columbia, 5358-2350 Health Sciences Mall, Vancouver, BC Canada V6T 1Z3
| | - Quin F. Wills
- Wellcome Trust Centre for Human Genetics, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - James D. Johnson
- Department of Cellular and Physiological Sciences, Faculty of Medicine, Diabetes Research Group, Life Sciences Institute, University of British Columbia, 5358-2350 Health Sciences Mall, Vancouver, BC Canada V6T 1Z3
| |
Collapse
|
25
|
Pronker MF, Bos TGAA, Sharp TH, Thies-Weesie DME, Janssen BJC. Olfactomedin-1 Has a V-shaped Disulfide-linked Tetrameric Structure. J Biol Chem 2015; 290:15092-101. [PMID: 25903135 PMCID: PMC4463452 DOI: 10.1074/jbc.m115.653485] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Indexed: 11/06/2022] Open
Abstract
Olfactomedin-1 (Olfm1; also known as noelin and pancortin) is a member of the olfactomedin domain-containing superfamily and a highly expressed neuronal glycoprotein important for nervous system development. It binds a number of secreted proteins and cell surface-bound receptors to induce cell signaling processes. Using a combined approach of x-ray crystallography, solution scattering, analytical ultracentrifugation, and electron microscopy we determined that full-length Olfm1 forms disulfide-linked tetramers with a distinctive V-shaped architecture. The base of the “V” is formed by two disulfide-linked dimeric N-terminal domains. Each of the two V legs consists of a parallel dimeric disulfide-linked coiled coil with a C-terminal β-propeller dimer at the tips. This agrees with our crystal structure of a C-terminal coiled-coil segment and β-propeller combination (Olfm1coil-Olf) that reveals a disulfide-linked dimeric arrangement with the β-propeller top faces in an outward exposed orientation. Similar to its family member myocilin, Olfm1 is stabilized by calcium. The dimer-of-dimers architecture suggests a role for Olfm1 in clustering receptors to regulate signaling and sheds light on the conformation of several other olfactomedin domain family members.
Collapse
Affiliation(s)
- Matti F Pronker
- From the Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research and
| | - Trusanne G A A Bos
- From the Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research and
| | - Thomas H Sharp
- Section Electron Microscopy, Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Dominique M E Thies-Weesie
- Van't Hoff Laboratory for Physical and Colloid Chemistry, Debye Institute of Nanomaterials Science, Department of Chemistry, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands and
| | - Bert J C Janssen
- From the Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research and
| |
Collapse
|
26
|
Li HM, Guo K, Yu Z, Feng R, Xu P. Diagnostic value of protein chips constructed by lung-cancer-associated markers selected by the T7 phage display library. Thorac Cancer 2015; 6:469-74. [PMID: 26273403 PMCID: PMC4511326 DOI: 10.1111/1759-7714.12215] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 11/05/2014] [Indexed: 12/17/2022] Open
Abstract
Background Traditional diagnostic technology with tumor biomarkers is inefficient, expensive and requires a large number of serum samples. The purpose of this study was to construct human lung cancer protein chips with new lung cancer biomarkers screened by the T7-phage display library, and improve the early diagnosis rate of lung cancer. Methods A T7-phage cDNA display library was constructed of fresh samples from 30 lung cancer patients. With biopanning and high-throughput screening, we gained the immunogenic phage clones from the cDNA library. The insert of selected phage was blasted at GeneBank for alignment to find the exact or the most similar known genes. Protein chips were then constructed and used to assay their expression level in lung cancer serum from 217 cases of lung cancer groups:80 cases of benign lung disease and 220 healthy controls. Results After four rounds of Biopanning and two rounds of enzyme-linked immunosorbent assay, 12 phage monoclonal samples were selected from 2880 phage monoclonal samples. After blasting at GeneBank, six similar genes were used to construct diagnostic protein chips. The protein chips were then used to assay expression level in lung cancer serum. The expression level of six genes in lung cancer groups was significantly higher than those in the other two groups (P < 0.05). Conclusions In this study, we successfully constructed diagnostic protein chips with biomarkers selected from the lung cancer T7-phage cDNA library, which can be used for the early screening of lung cancer patients.
Collapse
Affiliation(s)
- Hong-Mei Li
- Cancer Center, The Affiliated Hospital of Qingdao University Qingdao, China
| | - Kang Guo
- Cancer Center, The Affiliated Hospital of Qingdao University Qingdao, China
| | - Zhuang Yu
- Cancer Center, The Affiliated Hospital of Qingdao University Qingdao, China
| | - Rui Feng
- Cancer Center, The Affiliated Hospital of Qingdao University Qingdao, China
| | - Ping Xu
- Cancer Center, The Affiliated Hospital of Qingdao University Qingdao, China
| |
Collapse
|
27
|
Sultana A, Nakaya N, Dong L, Abu-Asab M, Qian H, Tomarev SI. Deletion of olfactomedin 2 induces changes in the AMPA receptor complex and impairs visual, olfactory, and motor functions in mice. Exp Neurol 2014; 261:802-11. [PMID: 25218043 DOI: 10.1016/j.expneurol.2014.09.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 08/21/2014] [Accepted: 09/02/2014] [Indexed: 12/19/2022]
Abstract
Olfactomedin 2 (Olfm2) is a secretory glycoprotein belonging to the family of olfactomedin domain-containing proteins. A previous study has shown that a mutation in OLFM2 is associated with primary open angle glaucoma in Japanese patients. In the present study, we generated Olfm2 deficient mice by replacing the Olfm2 gene with the LacZ gene. The loss of Olfm2 resulted in no gross abnormalities. However, Olfm2 null mice showed reduced exploration, locomotion, olfactory sensitivity, abnormal motor coordination, and anxiety related behavior. The pattern of the Olfm2 gene expression was studied in the brain and eye using β-galactosidase staining. In the brain, Olfm2 was mainly expressed in the olfactory bulb, cortex, piriform cortex, olfactory trabeculae, and inferior and superior colliculus. In the eye expression was detected mainly in retinal ganglion cells. In Olfm2 null mice, the amplitude of the first negative wave in the visual evoked potential test was significantly reduced as compared with wild-type littermates. Olfm2, similar to Olfm1, interacted with the GluR2 subunit of the AMPAR complexes and Olfm2 co-segregated with the AMPA receptor subunit GluR2 and other synaptic proteins in the synaptosomal membrane fraction upon biochemical fractionation of the adult mice cortex and retina. Immunoprecipitation from the synaptosomal membrane fraction of the Olfm2 null mouse brain cortex using the GluR2 antibody showed reduced levels of several components of the AMPAR complex in the immunoprecipitates including Olfm1, PSD95 and CNIH2. These results suggest that heterodimers of Olfm1 and Olfm2 interact with AMPAR more efficiently than Olfm2 homodimers and that Olfm2 plays a role in the organization of the AMPA receptor complexes.
Collapse
Affiliation(s)
- Afia Sultana
- Section on Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Naoki Nakaya
- Section on Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Lijin Dong
- Genetic Engineering Facility, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Mones Abu-Asab
- Histopathology Core Facility, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Haohua Qian
- Visual Function Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Stanislav I Tomarev
- Section on Retinal Ganglion Cell Biology, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
28
|
Anholt RRH. Olfactomedin proteins: central players in development and disease. Front Cell Dev Biol 2014; 2:6. [PMID: 25364714 PMCID: PMC4206993 DOI: 10.3389/fcell.2014.00006] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 02/07/2014] [Indexed: 12/14/2022] Open
Abstract
Olfactomedin proteins are characterized by a conserved domain of \texorpdfstring~\textasciitilde250 amino acids corresponding to the olfactomedin archetype first discovered in olfactory neuroepithelium. They arose early in evolution and occur throughout the animal kingdom. In mice and humans olfactomedin proteins comprise a diverse array of glycoproteins, many of which are critical for early development and functional organization of the nervous system as well as hematopoiesis. Olfactomedin domains appear to facilitate protein-protein interactions, intercellular interactions, and cell adhesion. Several members of the family have been implicated in various common diseases, notably myocilin in glaucoma and OLFM4 in cancer. This review highlights this important, hitherto understudied family of proteins.
Collapse
Affiliation(s)
- Robert R. H. Anholt
- Department of Biological Sciences and W. M. Keck Center for Behavioral Biology, North Carolina State UniversityRaleigh, NC, USA
| |
Collapse
|
29
|
Lu D, Dong W, Zhang X, Quan X, Bao D, Lu Y, Zhang L. WIF1 causes dysfunction of heart in transgenic mice. Transgenic Res 2013; 22:1179-89. [PMID: 23921644 PMCID: PMC3835953 DOI: 10.1007/s11248-013-9738-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 07/28/2013] [Indexed: 11/29/2022]
Abstract
Wnt activity is a key regulator of cardiac progenitor cell self-renewal, differentiation and morphogenesis. However, Wnt inhibitory factor 1 (WIF1), a antagonists of Wnt signaling activity, its potential effects on heart development has not yet been approached by either in vivo or in vitro studies. Here, the expression of WIF1 was regulated in a different way in the dilated and hypertrophic cardiomyopathy heart from transgenic mice by mutations in cardiac troponin T, cTnT(R141W) and cTnT(R92Q). The heart tissue specific transgenic mice of WIF1 was studied using M-mode echocardiography and histologic analyses. Production levels of an array of effectors and transcription factors that impact cellular organization and tissue morphology were measured. The effects of WIF1 on β-catenin pathway could be reversed by LiCl regarding signaling pathways and effector and respondent molecules in H9c2 cells, consistent with the expression levels of c-myc, natriuretic peptide precursor type B and skeletal muscle actin α1. Among the most noteworthy findings were that WIF1 impaired the function and structure of heart, and the effects on β-catenin pathway maybe the course of the former. It is anticipated that our findings will contribute to expansion of our understanding of WIF1 biological function on heart development and possible modes of treatment of heart diseases.
Collapse
Affiliation(s)
- Dan Lu
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Wei Dong
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Xu Zhang
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiongzhi Quan
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Dan Bao
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Yingdong Lu
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
| | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Beijing, People’s Republic of China
- Key Laboratory of Human Disease Animal Model, State Administration of Traditional Chinese Medicine, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing, 100021 People’s Republic of China
| |
Collapse
|
30
|
Deletion in the N-terminal half of olfactomedin 1 modifies its interaction with synaptic proteins and causes brain dystrophy and abnormal behavior in mice. Exp Neurol 2013; 250:205-18. [PMID: 24095980 DOI: 10.1016/j.expneurol.2013.09.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/16/2013] [Accepted: 09/19/2013] [Indexed: 11/24/2022]
Abstract
Olfactomedin 1 (Olfm1) is a secreted glycoprotein that is preferentially expressed in neuronal tissues. Here we show that deletion of exons 4 and 5 from the Olfm1 gene, which encodes a 52 amino acid long region in the N-terminal part of the protein, increased neonatal death and reduced body weight of surviving homozygous mice. Magnetic resonance imaging analyses revealed reduced brain volume and attenuated size of white matter tracts such as the anterior commissure, corpus callosum, and optic nerve. Adult Olfm1 mutant mice demonstrated abnormal behavior in several tests including reduced marble digging, elevated plus maze test, nesting activity and latency on balance beam tests as compared with their wild-type littermates. The olfactory system was both structurally and functionally disturbed by the mutation in the Olfm1 gene as shown by functional magnetic resonance imaging analysis and a smell test. Deficiencies of the olfactory system may contribute to the neonatal death and loss of body weight of Olfm1 mutant. Shotgun proteomics revealed 59 candidate proteins that co-precipitated with wild-type or mutant Olfm1 proteins in postnatal day 1 brain. Olfm1-binding targets included GluR2, Cav2.1, teneurin-4 and Kidins220. Modified interaction of Olfm1 with binding targets led to an increase in intracellular Ca(2+) concentration and activation of ERK1/2, MEK1 and CaMKII in the hippocampus and olfactory bulb of Olfm1 mutant mice compared with their wild-type littermates. Excessive activation of the CaMKII and Ras-ERK pathways in the Olfm1 mutant olfactory bulb and hippocampus by elevated intracellular calcium may contribute to the abnormal behavior and olfactory activity of Olfm1 mutant mice.
Collapse
|
31
|
Anholt RRH, Carbone MA. A molecular mechanism for glaucoma: endoplasmic reticulum stress and the unfolded protein response. Trends Mol Med 2013; 19:586-93. [PMID: 23876925 PMCID: PMC3795998 DOI: 10.1016/j.molmed.2013.06.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/20/2013] [Accepted: 06/28/2013] [Indexed: 10/26/2022]
Abstract
Primary open angle glaucoma (POAG) is a common late-onset neurodegenerative disease. Ocular hypertension represents a major risk factor, but POAG etiology remains poorly understood. Some cases of early-onset congenital glaucoma and adult POAG are linked to mutations in myocilin, a secreted protein of poorly defined function. Transgenic overexpression of myocilin in Drosophila and experiments in mice and human populations implicate the unfolded protein response (UPR) in the pathogenesis of glaucoma. We postulate that compromised ability of the UPR to eliminate misfolded mutant or damaged proteins, including myocilin, causes endoplasmic reticulum stress, resulting in functional impairment of trabecular meshwork cells that regulate intraocular pressure. This mechanism of POAG is reminiscent of other age-dependent neurodegenerative diseases that involve accumulation of protein aggregates.
Collapse
Affiliation(s)
- Robert R H Anholt
- Department of Biological Sciences and W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695-7617, USA.
| | | |
Collapse
|
32
|
Jin J, Cheng Y, Zhang Y, Wood W, Peng Q, Hutchison E, Mattson MP, Becker KG, Duan W. Interrogation of brain miRNA and mRNA expression profiles reveals a molecular regulatory network that is perturbed by mutant huntingtin. J Neurochem 2012; 123:477-90. [PMID: 22906125 PMCID: PMC3472040 DOI: 10.1111/j.1471-4159.2012.07925.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 08/13/2012] [Accepted: 08/14/2012] [Indexed: 11/27/2022]
Abstract
Emerging evidence indicates that microRNAs (miRNAs) may play an important role in the pathogenesis of Huntington's disease (HD). To identify the individual miRNAs that are altered in HD and may therefore regulate a gene network underlying mutant huntingtin-induced neuronal dysfunction in HD, we performed miRNA array analysis combined with mRNA profiling in the cerebral cortex from N171-82Q HD mice. Expression profiles of miRNAs as well as mRNAs in HD mouse cerebral cortex were analyzed and confirmed at different stages of disease progression; the most significant changes of miRNAs in the cerebral cortex were also detected in the striatum of HD mice. Our results revealed a significant alteration of miR-200 family members, miR-200a, and miR-200c in the cerebral cortex and the striatum, at the early stage of disease progression in N171-82Q HD mice. We used a coordinated approach to integrate miRNA and mRNA profiling, and applied bioinformatics to predict a target gene network potentially regulated by these significantly altered miRNAs that might be involved in HD disease progression. Interestingly, miR-200a and miR-200c are predicted to target genes regulating synaptic function, neurodevelopment, and neuronal survival. Our results suggest that altered expression of miR-200a and miR-200c may interrupt the production of proteins involved in neuronal plasticity and survival, and further investigation of the involvement of perturbed miRNA expression in HD pathogenesis is warranted, and may lead to reveal novel approaches for HD therapy.
Collapse
Affiliation(s)
- Jing Jin
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Yong Cheng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Yongqing Zhang
- Gene Expression and Genomics Unit Research Resources Branch, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
| | - William Wood
- Gene Expression and Genomics Unit Research Resources Branch, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
| | - Qi Peng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Emmette Hutchison
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Kevin G. Becker
- Gene Expression and Genomics Unit Research Resources Branch, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| |
Collapse
|
33
|
Sánchez-Hernández D, Sierra J, Ortigão-Farias JR, Guerrero I. The WIF domain of the human and Drosophila Wif-1 secreted factors confers specificity for Wnt or Hedgehog. Development 2012; 139:3849-58. [PMID: 22951645 DOI: 10.1242/dev.080028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The Hedgehog (Hh) and Wnt signaling pathways are crucial for development as well as for adult stem cell maintenance in all organisms from Drosophila to humans. Aberrant activation of these pathways has been implicated in many types of human cancer. During evolution, organisms have developed numerous ways to fine-tune Wnt and Hh signaling. One way is through extracellular modulators that directly interact with Wnt or Hh, such as the Wnt inhibitory factor (Wif-1) family of secreted factors. Interestingly, Wif-1 family members have divergent functions in the Wnt and Hh pathways in different organisms. Whereas vertebrate Wif-1 blocks Wnt signaling, Drosophila Wif-1 [Shifted (Shf)] regulates only Hh distribution and spreading through the extracellular matrix. Here, we investigate which parts of the Shf and human Wif-1 (WIF1) proteins are responsible for functional divergence. We analyze the behavior of domain-swap (the Drosophila and human WIF domain and EGF repeats) chimeric constructs during wing development. We demonstrate that the WIF domain confers the specificity for Hh or Wg morphogen. The EGF repeats are important for the interaction of Wif-1 proteins with the extracellular matrix; Drosophila EGF repeats preferentially interact with the glypican Dally-like (Dlp) when the WIF domain belongs to human WIF1 and with Dally when the WIF domain comes from Shf. These results are important both from the evolutionary perspective and for understanding the mechanisms of morphogen distribution in a morphogenetic field.
Collapse
Affiliation(s)
- David Sánchez-Hernández
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Cantoblanco, E-28049 Madrid, Spain
| | | | | | | |
Collapse
|
34
|
Nakaya N, Sultana A, Lee HS, Tomarev SI. Olfactomedin 1 interacts with the Nogo A receptor complex to regulate axon growth. J Biol Chem 2012; 287:37171-84. [PMID: 22923615 DOI: 10.1074/jbc.m112.389916] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Olfm1, a secreted highly conserved glycoprotein, is detected in peripheral and central nervous tissues and participates in neural progenitor maintenance, cell death in brain, and optic nerve arborization. In this study, we identified Olfm1 as a molecule promoting axon growth through interaction with the Nogo A receptor (NgR1) complex. Olfm1 is coexpressed with NgR1 in dorsal root ganglia and retinal ganglion cells in embryonic and postnatal mice. Olfm1 specifically binds to NgR1, as judged by alkaline phosphatase assay and coimmunoprecipitation. The addition of Olfm1 inhibited the growth cone collapse of dorsal root ganglia neurons induced by myelin-associated inhibitors, indicating that Olfm1 attenuates the NgR1 receptor functions. Olfm1 caused the inhibition of NgR1 signaling by interfering with interaction between NgR1 and its coreceptors p75NTR or LINGO-1. In zebrafish, inhibition of optic nerve extension by olfm1 morpholino oligonucleotides was partially rescued by dominant negative ngr1 or lingo-1. These data introduce Olfm1 as a novel NgR1 ligand that may modulate the functions of the NgR1 complex in axonal growth.
Collapse
Affiliation(s)
- Naoki Nakaya
- Section of Molecular Mechanisms of Glaucoma, Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892-0606, USA
| | | | | | | |
Collapse
|
35
|
Keenan J, Joyce H, Aherne S, O'Dea S, Doolan P, Lynch V, Clynes M. Olfactomedin III expression contributes to anoikis-resistance in clonal variants of a human lung squamous carcinoma cell line. Exp Cell Res 2012; 318:593-602. [DOI: 10.1016/j.yexcr.2012.01.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 12/09/2011] [Accepted: 01/07/2012] [Indexed: 10/14/2022]
|
36
|
The role of glypicans in Wnt inhibitory factor-1 activity and the structural basis of Wif1's effects on Wnt and Hedgehog signaling. PLoS Genet 2012; 8:e1002503. [PMID: 22383891 PMCID: PMC3285576 DOI: 10.1371/journal.pgen.1002503] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 12/11/2011] [Indexed: 01/03/2023] Open
Abstract
Proper assignment of cellular fates relies on correct interpretation of Wnt and Hedgehog (Hh) signals. Members of the Wnt Inhibitory Factor-1 (WIF1) family are secreted modulators of these extracellular signaling pathways. Vertebrate WIF1 binds Wnts and inhibits their signaling, but its Drosophila melanogaster ortholog Shifted (Shf) binds Hh and extends the range of Hh activity in the developing D. melanogaster wing. Shf activity is thought to depend on reinforcing interactions between Hh and glypican HSPGs. Using zebrafish embryos and the heterologous system provided by D. melanogaster wing, we report on the contribution of glypican HSPGs to the Wnt-inhibiting activity of zebrafish Wif1 and on the protein domains responsible for the differences in Wif1 and Shf specificity. We show that Wif1 strengthens interactions between Wnt and glypicans, modulating the biphasic action of glypicans towards Wnt inhibition; conversely, glypicans and the glypican-binding “EGF-like” domains of Wif1 are required for Wif1's full Wnt-inhibiting activity. Chimeric constructs between Wif1 and Shf were used to investigate their specificities for Wnt and Hh signaling. Full Wnt inhibition required the “WIF” domain of Wif1, and the HSPG-binding EGF-like domains of either Wif1 or Shf. Full promotion of Hh signaling requires both the EGF-like domains of Shf and the WIF domains of either Wif1 or Shf. That the Wif1 WIF domain can increase the Hh promoting activity of Shf's EGF domains suggests it is capable of interacting with Hh. In fact, full-length Wif1 affected distribution and signaling of Hh in D. melanogaster, albeit weakly, suggesting a possible role for Wif1 as a modulator of vertebrate Hh signaling. In developing organisms, cells choose between alternative fates in order to make appropriately patterned tissues, and misregulation of those choices can underlie both developmental defects and cancers. Cells often make these decisions because of signals received from neighboring cells, such as those mediated by the secreted signaling proteins of the Wnt and Hedgehog (Hh) families. While signaling can be regulated by the levels of signaling or receptor proteins expressed by cells, another level of control is exerted by proteins that bind signaling proteins outside of cells and either inhibit or promote the signaling process. In the fruitfly Drosophilamelanogaster, the secreted Shifted protein has been shown to bind Hh and to increase Hh signaling, likely by reinforcing interactions between Hh and cell surface proteins of the glypican family. We provide evidence that the vertebrate homolog of Shifted, Wnt Inhibitory Factor-1 (Wif1), inhibits Wnt activity by a similar mechanism, reinforcing interactions between Wnts and glypicans in a manner that sequesters Wnts from their receptors. We also examine the structural basis for the specificities of Wif1 and Shifted for Wnt and Hh signaling, respectively, and provide evidence that Wif1, although a potent inhibitor of Wnt activity, influences D. melanogaster Hh signaling.
Collapse
|
37
|
Wnt activation downregulates olfactomedin-1 in Fallopian tubal epithelial cells: a microenvironment predisposed to tubal ectopic pregnancy. J Transl Med 2012; 92:256-64. [PMID: 21968811 PMCID: PMC3272473 DOI: 10.1038/labinvest.2011.148] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Ectopic pregnancy (EP) occurs when the embryo fails to transit to the uterus and attach to the luminal epithelium of the Fallopian tube (FT). Tubal EP is a common gynecological emergency and more than 95% of EP occurs in the ampullary region of the FT. In humans, Wnt activation and downregulation of olfactomedin-1 (Olfm-1) occur in the receptive endometrium and coincided with embryo implantation in vivo. Whether similar molecular changes happen in the FT leading to EP remains unclear. We hypothesized that activation of Wnt signaling downregulates Olfm-1 expression predisposes to EP. We investigated the spatiotemporal expression of Olfm-1 in FT from non-pregnant women and women with EP, and used a novel trophoblastic spheroid (embryo surrogate)-FT epithelial cell co-culture model (JAr and OE-E6/E7 cells) to study the role of Olfm-1 on spheroid attachment. Olfm-1 mRNA expression in the ampullary region of non-pregnant FT was higher (P<0.05) in the follicular phase than in the luteal phase. Ampullary tubal Olfm-1 expression was lower in FT from women with EP compared to normal controls at the luteal phase (histological scoring (H-SCORE)=1.3±0.2 vs 2.4±0.5; P<0.05). Treatment of OE-E6/E7 with recombinant Olfm-1 (0.2-5 μg/ml) suppressed spheroid attachment to OE-E6/E7 cells, while activation of Wnt-signaling pathway by Wnt3a or LiCl reduced endogenous Olfm-1 expression and increased spheroid attachment. Conversely, suppression of Olfm-1 expression by RNAi increased spheroid attachment to OE-E6/E7 cells. Taken together, Wnt activation suppresses Olfm-1 expression, and this may predispose a favorable microenvironment of the retained embryo in the FT, leading to EP in humans.
Collapse
|
38
|
Schiro MM, Stauber SE, Peterson TL, Krueger C, Darnell SJ, Satyshur KA, Drinkwater NR, Newton MA, Hoffmann FM. Mutations in protein-binding hot-spots on the hub protein Smad3 differentially affect its protein interactions and Smad3-regulated gene expression. PLoS One 2011; 6:e25021. [PMID: 21949838 PMCID: PMC3176292 DOI: 10.1371/journal.pone.0025021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 08/22/2011] [Indexed: 02/06/2023] Open
Abstract
Background Hub proteins are connected through binding interactions to many other proteins. Smad3, a mediator of signal transduction induced by transforming growth factor beta (TGF-β), serves as a hub protein for over 50 protein-protein interactions. Different cellular responses mediated by Smad3 are the product of cell-type and context dependent Smad3-nucleated protein complexes acting in concert. Our hypothesis is that perturbation of this spectrum of protein complexes by mutation of single protein-binding hot-spots on Smad3 will have distinct consequences on Smad3-mediated responses. Methodology/Principal Findings We mutated 28 amino acids on the surface of the Smad3 MH2 domain and identified 22 Smad3 variants with reduced binding to subsets of 17 Smad3-binding proteins including Smad4, SARA, Ski, Smurf2 and SIP1. Mutations defective in binding to Smad4, e.g., D408H, or defective in nucleocytoplasmic shuttling, e.g., W406A, were compromised in modulating the expression levels of a Smad3-dependent reporter gene or six endogenous Smad3-responsive genes: Mmp9, IL11, Tnfaip6, Fermt1, Olfm2 and Wnt11. However, the Smad3 mutants Y226A, Y297A, W326A, K341A, and E267A had distinct differences on TGF-β signaling. For example, K341A and Y226A both reduced the Smad3-mediated activation of the reporter gene by ∼50% but K341A only reduced the TGF-β inducibilty of Olfm2 in contrast to Y226A which reduced the TGF-β inducibility of all six endogenous genes as severely as the W406A mutation. E267A had increased protein binding but reduced TGF-β inducibility because it caused higher basal levels of expression. Y297A had increased TGF-β inducibility because it caused lower Smad3-induced basal levels of gene expression. Conclusions/Significance Mutations in protein binding hot-spots on Smad3 reduced the binding to different subsets of interacting proteins and caused a range of quantitative changes in the expression of genes induced by Smad3. This approach should be useful for unraveling which Smad3 protein complexes are critical for specific biological responses.
Collapse
Affiliation(s)
- Michelle M. Schiro
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Sara E. Stauber
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Tami L. Peterson
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Chateen Krueger
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Steven J. Darnell
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Kenneth A. Satyshur
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Norman R. Drinkwater
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Michael A. Newton
- Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
| | - F. Michael Hoffmann
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
39
|
Malinauskas T, Aricescu AR, Lu W, Siebold C, Jones EY. Modular mechanism of Wnt signaling inhibition by Wnt inhibitory factor 1. Nat Struct Mol Biol 2011; 18:886-93. [PMID: 21743455 DOI: 10.1038/nsmb.2081] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 05/06/2011] [Indexed: 01/12/2023]
Abstract
Wnt morphogens control embryonic development and homeostasis in adult tissues. In vertebrates the N-terminal WIF domain (WIF-1(WD)) of Wnt inhibitory factor 1 (WIF-1) binds Wnt ligands. Our crystal structure of WIF-1(WD) reveals a previously unidentified binding site for phospholipid; two acyl chains extend deep into the domain, and the head group is exposed to the surface. Biophysical and cellular assays indicate that there is a WIF-1(WD) Wnt-binding surface proximal to the lipid head group but also implicate the five epidermal growth factor (EGF)-like domains (EGFs I-V) in Wnt binding. The six-domain WIF-1 crystal structure shows that EGFs I-V are wrapped back, interfacing with WIF-1(WD) at EGF III. EGFs II-V contain a heparan sulfate proteoglycan (HSPG)-binding site, consistent with conserved positively charged residues on EGF IV. This combination of HSPG- and Wnt-binding properties suggests a modular model for the localization of WIF-1 and for signal inhibition within morphogen gradients.
Collapse
Affiliation(s)
- Tomas Malinauskas
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | | | | | | |
Collapse
|
40
|
Menaa F, Braghini CA, Vasconcellos JPCD, Menaa B, Costa VP, Figueiredo ESD, Melo MBD. Keeping an eye on myocilin: a complex molecule associated with primary open-angle glaucoma susceptibility. Molecules 2011; 16:5402-21. [PMID: 21709622 PMCID: PMC6264709 DOI: 10.3390/molecules16075402] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 06/21/2011] [Indexed: 02/04/2023] Open
Abstract
MYOC encodes a secretary glycoprotein of 504 amino acids named myocilin. MYOC is the first gene to be linked to juvenile open-angle glaucoma (JOAG) and some forms of adult-onset primary open-angle glaucoma (POAG). The gene was identified as an up-regulated molecule in cultured trabecular meshwork (TM) cells after treatment with dexamethasone and was originally referred to as trabecular meshwork-inducible glucocorticoid response (TIGR). Elevated intraocular pressure (IOP), due to decreased aqueous outflow, is the strongest known risk factor for POAG. Increasing evidence showed that the modulation of the wild-type (wt) myocilin protein expression is not causative of glaucoma while some misfolded and self-assembly aggregates of mutated myocilin may be associated with POAG in related or unrelated populations. The etiology of the disease remains unclear. Consequently, a better understanding of the molecular mechanisms underlyingPOAG is required to obtain early diagnosis, avoid potential disease progression, and develop new therapeutic strategies. In the present study, we review and discuss the most relevant studies regarding structural characterizations, expressions, molecular interactions, putative functions of MYOC gene and/or its corresponding protein in POAG etiology.
Collapse
Affiliation(s)
- Farid Menaa
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Campinas-SP 13083-875, Brazil; (C.A.B.); (M.B.D.M.)
- Author to whom correspondence should be addressed; ; or ; Tel.: +55-19-3521-1138; Fax: +55-19-3521-1089
| | - Carolina Ayumi Braghini
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Campinas-SP 13083-875, Brazil; (C.A.B.); (M.B.D.M.)
| | - Jose Paulo Cabral De Vasconcellos
- Department of Ophthalmology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas-SP 13083-888, Brazil; (J.P.C.D.V.); (V.P.C.); (E.S.D.F.)
| | - Bouzid Menaa
- Department of Chemistry and Nanobiotechnology, Fluorotronics, Inc., San Diego, CA 92081, USA; (B.M.)
| | - Vital Paulino Costa
- Department of Ophthalmology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas-SP 13083-888, Brazil; (J.P.C.D.V.); (V.P.C.); (E.S.D.F.)
| | - Eugênio Santana De Figueiredo
- Department of Ophthalmology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas-SP 13083-888, Brazil; (J.P.C.D.V.); (V.P.C.); (E.S.D.F.)
| | - Monica Barbosa De Melo
- Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Campinas-SP 13083-875, Brazil; (C.A.B.); (M.B.D.M.)
| |
Collapse
|
41
|
Sultana A, Nakaya N, Senatorov VV, Tomarev SI. Olfactomedin 2: expression in the eye and interaction with other olfactomedin domain-containing proteins. Invest Ophthalmol Vis Sci 2011; 52:2584-92. [PMID: 21228389 DOI: 10.1167/iovs.10-6356] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Olfactomedin 2 (OLFM2) belongs to the family of olfactomedin domain-containing proteins. Genetic data suggest its association with glaucoma in Japanese patients. However, its functions are still elusive. In this study, the properties of mammalian OLFM2 were investigated. METHODS Expression of the rat and mouse Olfm2 gene was studied by using real-time PCR and in situ hybridization. Substitutions were introduced into OLFM2 by mutagenesis in vitro. Intracellular localization of OLFM2 was studied by confocal microscopy after transient transfection in HEK293 cells. Interaction of OLFM2 with olfactomedin 1 (Olfm1), olfactomedin 3 (Olfm3), myocilin, and gliomedin was studied by using co-immunoprecipitation. RESULTS Two major human OLFM2 mRNAs encode secreted proteins with a length of 454 and 478 amino acids. OLFM2 is more closely related to OLFM1 and -3 than to any other family members. Olfm2 showed the most dynamic expression pattern compared with Olfm1 and -3 during mouse eye development and was expressed preferentially in the developing retinal ganglion cell layer. Among three OLFM2 substitutions tested (T86M, R144Q, and L420S), only L420S completely blocked secretion of the protein. OLFM2 interacted with Olfm1 and -3, but not with myocilin and gliomedin. Co-transfection of the L420S mutant with wild-type Olfm1 and -3 significantly inhibited secretion of Olfm1 and -3. CONCLUSIONS Highly conserved OLFM2 protein may play an important role in the course of retinal and eye development. Severe mutations in one of the closely related olfactomedin domain-containing proteins (Olfm1-3) may block the secretion and probably the activity of all three family members, leading to more pronounced diseases of the retina than the knockout of individual genes.
Collapse
Affiliation(s)
- Afia Sultana
- Section of Molecular Mechanisms of Glaucoma, Laboratory of Molecular and Developmental Biology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
42
|
Grover PK, Hardingham JE, Cummins AG. Stem cell marker olfactomedin 4: critical appraisal of its characteristics and role in tumorigenesis. Cancer Metastasis Rev 2011; 29:761-75. [PMID: 20878207 DOI: 10.1007/s10555-010-9262-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Olfactomedin 4 (OLFM4), a member of the olfactomedin domain-containing proteins, is a glycoprotein with molecular weight of approximately 64 kDa. The protein is a "robust marker" of Lgr5+ stem cells and has been localised to mitochondria, nuclei and cell membranes. The bulk of OLFM4 exists in a polymeric form which is held together by disulfide bonds and carbohydrate interactions. Earlier studies revealed that the protein binds to lectins and cadherins, and facilitates cell-cell adhesion. Recent data demonstrated that the protein possesses several hallmarks of carcinogenesis. OLFM4 has also been purported to be an inducible resistance factor to apoptotic stimuli such as radiation and anticancer drugs. Here, we review its synonyms and classification, gene structure, protein structure, intracellular and tissue distribution, adhesive and antiapoptotic; mitotic; migratory and cell cycle regulatory characteristics. We also critically evaluate recent advances in understanding of the transcriptional regulation of OLFM4 and its upstream signalling pathways with special emphasis on carcinogenesis and outline future perspectives in the field.
Collapse
Affiliation(s)
- Phulwinder K Grover
- Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, 28 Woodville Road, Woodville South, South Australia 5011, Australia.
| | | | | |
Collapse
|
43
|
Kodithuwakku SP, Ng PY, Liu Y, Ng EHY, Yeung WSB, Ho PC, Lee KF. Hormonal regulation of endometrial olfactomedin expression and its suppressive effect on spheroid attachment onto endometrial epithelial cells. Hum Reprod 2010; 26:167-75. [DOI: 10.1093/humrep/deq298] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
44
|
Bohr DC, Koch M, Kritzenberger M, Fuchshofer R, Tamm ER. Increased expression of olfactomedin-1 and myocilin in podocytes during puromycin aminonucleoside nephrosis. Nephrol Dial Transplant 2010; 26:83-92. [PMID: 20595200 DOI: 10.1093/ndt/gfq366] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The olfactomedin domain proteins Olfm-1 and myocilin are expressed in podocytes. Myocilin stimulates the formation of focal contacts and actin stress fibres in podocytes and other cell types, effects that are mediated through the Wnt signalling pathway. Here, we tested if the expression of both proteins is modified during puromycin aminonucleoside (PAN) nephrosis, which leads to structural changes in the actin cytoskeleton of podocytes. METHODS Rats were treated with PAN, and the effectiveness of treatment was analysed by electron microscopy of podocytes and protein detection in the urine. The expression of Olfm-1 and myocilin was studied by immunohistochemistry, western blot analysis of glomerular proteins and real-time RT-PCR of glomerular proteins. In parallel experiments, the expression of Olfm-1 was studied in cultured podocytes treated with dexamethasone, TGF-β, TNF-α and PAN. RESULTS Between Days 5 and 22 after treatment, the amounts of the BMZ and BMY splice variants of Olfm-1 and their mRNA were markedly elevated in proteins and mRNA from isolated glomeruli. Immunohistochemistry showed that the expression of Olfm-1 was confined to podocytes. Essentially, comparable results were obtained for myocilin. The BMZ variant of Olfm-1 appeared to be secreted from podocytes and was found in high amounts in urine of treated animals. Treatment of cultured podocytes with dexamethasone and PAN caused an increase in Olfm-1 expression, while treatment with recombinant Olfm-1 increased the formation of actin stress fibres. CONCLUSIONS Olfm-1 and myocilin are markedly induced in podocytes during PAN nephrosis and appear to be involved in the processes that govern the reorganization of the actin cytoskeleton during podocyte repair.
Collapse
Affiliation(s)
- Daniela C Bohr
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | | | | | | | | |
Collapse
|
45
|
Yilmaz G, Alexander JS, Erkuran Yilmaz C, Granger DN. Induction of neuro-protective/regenerative genes in stem cells infiltrating post-ischemic brain tissue. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2010; 2:11. [PMID: 20509949 PMCID: PMC2893124 DOI: 10.1186/2040-7378-2-11] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 05/28/2010] [Indexed: 12/25/2022]
Abstract
BACKGROUND Although the therapeutic potential of bone marrow-derived stromal stem cells (BMSC) has been demonstrated in different experimental models of ischemic stroke, it remains unclear how stem cells (SC) induce neuroprotection following stroke. In this study, we describe a novel method for isolating BMSC that infiltrate postischemic brain tissue and use this method to identify the genes that are persistently activated or depressed in BMSC that infiltrate brain tissue following ischemic stroke. METHODS Ischemic strokes were induced in C57BL/6 mice by middle cerebral artery occlusion for 1 h, followed by reperfusion. BMSC were isolated from H-2 Kb-tsA58 (immortomouse) mice, and were administered (i.v.) 24 h after reperfusion. At the peak of therapeutic improvement (14 days after the ischemic insult), infarcted brain tissue was isolated, and the BMSC were isolated by culturing at 33 degrees C. Microarray analysis and RT-PCR were performed to compare differential gene expression between naïve and infiltrating BMSC populations. RESULTS Z-scoring revealed dramatic differences in the expression of extracellular genes between naïve and infiltrating BMSC. Pair-wise analysis detected 80 extracellular factor genes that were up-regulated (>/= 2 fold, P < 0.05, Benjamini-Hochberg correction) between naïve and infiltrated BMSC. Although several anticipated neuroregenerative, nerve guidance and angiogenic factor (e.g., bFGF, bone morphogenetic protein, angiopoietins, neural growth factor) genes exhibited an increased expression, a remarkable induction of genes for nerve guidance survival (e.g., cytokine receptor-like factor 1, glypican 1, Dickkopf homolog 2, osteopontin) was also noted. CONCLUSIONS BMSC infiltrating the post-ischemic brain exhibit persistent epigenetic changes in gene expression for numerous extracellular genes, compared to their naïve counterparts. These genes are relevant to the neuroprotection, regeneration and angiogenesis previously described following stem cell therapy in animal models of ischemic stroke.
Collapse
Affiliation(s)
- Gokhan Yilmaz
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA.
| | | | | | | |
Collapse
|
46
|
Chen CM, Chiu SL, Shen W, Cline HT. Co-expression of Argonaute2 Enhances Short Hairpin RNA-induced RNA Interference in Xenopus CNS Neurons In Vivo. Front Neurosci 2009; 3:63. [PMID: 20582287 PMCID: PMC2858607 DOI: 10.3389/neuro.17.001.2009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Accepted: 06/16/2009] [Indexed: 11/25/2022] Open
Abstract
RNA interference (RNAi) is an evolutionarily conserved mechanism for sequence-specific gene silencing. Recent advances in our understanding of RNAi machinery make it possible to reduce protein expression by introducing short hairpin RNA (shRNA) into cells of many systems, however, the efficacy of RNAi-mediated protein knockdown can be quite variable, especially in intact animals, and this limits its application. We built adaptable molecular tools, pSilencer (pSi) and pReporter (pRe) constructs, to evaluate the impact of different promoters, shRNA structures and overexpression of Ago2, the key enzyme in the RNA-induced silencing complex, on the efficiency of RNAi. The magnitude of RNAi knockdown was evaluated in cultured cells and intact animals by comparing fluorescence intensity levels of GFP, the RNAi target, relative to mCherry, which was not targeted. Co-expression of human Ago2 with shRNA significantly enhanced efficiency of GFP knockdown in cell lines and in neurons of intact Xenopus tadpoles. Human H1- and U6-promotors alone or the U6-promotor with an enhancer element were equally effective at driving GFP knockdown. shRNA derived from the microRNA-30 design (shRNAmir30) enhanced the efficiency of GFP knockdown. Expressing pSi containing Ago2 with shRNA increased knockdown efficiency of an endogenous neuronal protein, the GluR2 subunit of the AMPA receptor, functionally accessed by recording AMPA receptor-mediated spontaneous synaptic currents in Xenopus CNS neurons. Our data suggest that co-expression of Ago2 and shRNA is a simple method to enhance RNAi in intact animals. While morpholino antisense knockdown is effective in Xenopus and Zebrafish, a principle advantage of the RNAi method is the possibility of spatial and temporal control of protein knockdown by use of cell type specific and regulatable pol II promoters to drive shRNA and Ago2. This should extend the application of RNAi to study gene function of intact brain circuits.
Collapse
Affiliation(s)
- Chih-Ming Chen
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory Cold Spring Harbor, NY, USA
| | | | | | | |
Collapse
|
47
|
Abstract
It is well documented that mutations in the MYOCILIN gene may lead to juvenile- and adult-onset primary open-angle glaucoma. However, the functions of wild-type myocilin are still not well understood. To study the functions of human myocilin and its two proteolytic fragments, these proteins were expressed in HEK293 cells. Conditioned medium from myocilin-expressing cells, as well as purified myocilin, induced the formation of stress fibers in primary cultures of human trabecular meshwork or NIH 3T3 cells. Stress fiber-inducing activity of myocilin was blocked by antibodies against myocilin, as well as secreted inhibitors of Wnt signaling, secreted Frizzled-related protein 1 (sFRP1) or sFRP3, and beta-catenin small interfering RNA. Interaction of myocilin with sFRP1, sFRP3, and several Frizzled receptors was confirmed by immunoprecipitation experiments and by binding of myocilin to the surface of cells expressing cysteine-rich domains of different Frizzled and sFRPs. Treatment of NIH 3T3 cells with myocilin and its fragments induced intracellular redistribution of beta-catenin and its accumulation on the cellular membrane but did not induce nuclear accumulation of beta-catenin. Overexpression of myocilin in the eye angle tissues of transgenic mice stimulated accumulation of beta-catenin in these tissues. Myocilin and Wnt proteins may perform redundant functions in the mammalian eye, since myocilin modulates Wnt signaling by interacting with components of this signaling pathway.
Collapse
|
48
|
Cyclops. Can J Ophthalmol 2008. [DOI: 10.3129/i08-125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
49
|
Recent Papers on Zebrafish and Other Aquarium Fish Models. Zebrafish 2008. [DOI: 10.1089/zeb.2008.9987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
50
|
Tomarev SI, Nakaya N. In-vitro study of the activity of ciprofloxacin alone and in combination against strains of Pseudomonas aeruginosa with multiple antibiotic resistance. J Antimicrob Chemother 1986; 40:122-38. [PMID: 19554483 DOI: 10.1007/s12035-009-8076-x] [Citation(s) in RCA: 123] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Accepted: 06/14/2009] [Indexed: 01/25/2023] Open
Abstract
Ciprofloxacin appears to have useful activity against Pseudomonas aeruginosa. We have studied its in-vitro activity against ten strains of Ps. aeruginosa with multiple antibiotic resistance. We have confirmed that ciprofloxacin is very active against Ps. aeruginosa with minimal inhibitory concentrations ranging from 0.07 to 0.7 mg/l. Killing curves show ciprofloxacin to be rapidly bactericidal with no regrowth after 24 h. Checkerboard studies with ciprofloxacin in combination with gentamicin, azlocillin and ceftazidime show no consistent interaction. These studies suggest that ciprofloxacin should prove a useful antibiotic in treating infections caused by multiresistant Ps. aeruginosa.
Collapse
Affiliation(s)
- Stanislav I Tomarev
- Section of Molecular Mechanisms of Glaucoma, Laboratory of Molecular and Developmental Biology, National Eye Institute, NIH, 5635 Fishers Lane, Room 1124, Bethesda, MD, 20892, USA.
| | | |
Collapse
|