1
|
Suzuki Y, Emoto T, Sato S, Yoshida T, Shoda M, Endoh H, Nagao M, Hamana T, Inoue T, Hayashi T, Nitta E, Konishi H, Kiuchi K, Takami M, Imamura K, Taniguchi M, Inoue M, Nakamura T, Sonoda Y, Takahara H, Nakasone K, Yamamoto K, Tani K, Iwai H, Nakanishi Y, Yonehara S, Murakami A, Toh R, Ohkawa T, Furuyashiki T, Nitta R, Yamashita T, Hirata KI, Fukuzawa K. Left atrial single-cell transcriptomics reveals amphiregulin as a surrogate marker for atrial fibrillation. Commun Biol 2024; 7:1601. [PMID: 39622943 PMCID: PMC11612213 DOI: 10.1038/s42003-024-07308-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
Atrial fibrillation (AF) is strongly associated with strokes, heart failure, and increased mortality. This study aims to identify the monocyte-macrophage heterogeneity and interactions of these cells with non-immune cells, and to identify functional biomarkers in patients with AF. Therefore, we assess the single cell landscape of left atria (LA), using a combination of single cell and nucleus RNA-seq. Myeloid cells in LA tissue are categorized into five macrophage clusters, three monocyte clusters, and others. Cell-Chat analysis revealed that monocytes and IL1B+ macrophages send epidermal growth factor (EGF) signals to fibroblasts. Amphiregulin (AREG) is the most upregulated gene in monocytes and IL1B+ macrophages in the AF group, compared with healthy controls from other groups. Serum AREG levels are higher in patients with persistent AF. These data suggested that EGF signaling pathway could be a therapeutic target for AF and serum AREG levels provide an effective biomarker for predicting persistent AF.
Collapse
Affiliation(s)
- Yuya Suzuki
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takuo Emoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Shunsuke Sato
- Division of Cardiovascular Surgery, Department of Surgery, Yodogawa Christian Hospital, Osaka, Japan
| | - Takeshi Yoshida
- Department of Information and Intelligence Engineering, Kobe University, Kobe, Japan
| | - Mitsuhiko Shoda
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiromi Endoh
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Manabu Nagao
- Division of Evidence-Based Laboratory Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyo Hamana
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Taishi Inoue
- Division of Cardiovascular Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomohiro Hayashi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Eriko Nitta
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroki Konishi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Yodogawa Christian Hospital, Osaka, Japan
| | - Kunihiko Kiuchi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Mitsuru Takami
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kimitake Imamura
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Section of Arrhythmia, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masayuki Taniguchi
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masatoshi Inoue
- Department of Information and Intelligence Engineering, Kobe University, Kobe, Japan
| | - Toshihiro Nakamura
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yusuke Sonoda
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroyuki Takahara
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazutaka Nakasone
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kyoko Yamamoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenichi Tani
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hidehiro Iwai
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yusuke Nakanishi
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shogo Yonehara
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Atsushi Murakami
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryuji Toh
- Division of Evidence-Based Laboratory Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takenao Ohkawa
- Department of Information and Intelligence Engineering, Kobe University, Kobe, Japan
| | - Tomoyuki Furuyashiki
- Division of Pharmacology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryo Nitta
- Division of Structural Medicine and Anatomy, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoya Yamashita
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Advanced Medical Science, Technology and Innovation, Kobe University Graduate School of Science, Kobe, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koji Fukuzawa
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Section of Arrhythmia, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
2
|
Erickson MA, Johnson RS, Damodarasamy M, MacCoss MJ, Keene CD, Banks WA, Reed MJ. Data-independent acquisition proteomic analysis of the brain microvasculature in Alzheimer's disease identifies major pathways of dysfunction and upregulation of cytoprotective responses. Fluids Barriers CNS 2024; 21:84. [PMID: 39434151 PMCID: PMC11492478 DOI: 10.1186/s12987-024-00581-1] [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: 08/29/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024] Open
Abstract
Brain microvascular dysfunction is an important feature of Alzheimer's disease (AD). To better understand the brain microvascular molecular signatures of AD, we processed and analyzed isolated human brain microvessels by data-independent acquisition liquid chromatography with tandem mass spectrometry (DIA LC-MS/MS) to generate a quantitative dataset at the peptide and protein level. Brain microvessels were isolated from parietal cortex grey matter using protocols that preserve viability for downstream functional studies. Our cohort included 23 subjects with clinical and neuropathologic concordance for Alzheimer's disease, and 21 age-matched controls. In our analysis, we identified 168 proteins whose abundance was significantly increased, and no proteins that were significantly decreased in AD. The most highly increased proteins included amyloid beta, tau, midkine, SPARC related modular calcium binding 1 (SMOC1), and fatty acid binding protein 7 (FABP7). Additionally, Gene Ontology (GO) enrichment analysis identified the enrichment of increased proteins involved in cellular detoxification and antioxidative responses. A systematic evaluation of protein functions using the UniProt database identified groupings into common functional themes including the regulation of cellular proliferation, cellular differentiation and survival, inflammation, extracellular matrix, cell stress responses, metabolism, coagulation and heme breakdown, protein degradation, cytoskeleton, subcellular trafficking, cell motility, and cell signaling. This suggests that AD brain microvessels exist in a stressed state of increased energy demand, and mount a compensatory response to ongoing oxidative and cellular damage that is associated with AD. We also used public RNAseq databases to identify cell-type enriched genes that were detected at the protein level and found no changes in abundance of these proteins between control and AD groups, indicating that changes in cellular composition of the isolated microvessels were minimal between AD and no-AD groups. Using public data, we additionally found that under half of the proteins that were significantly increased in AD microvessels had concordant changes in brain microvascular mRNA, implying substantial discordance between gene and protein levels. Together, our results offer novel insights into the molecular underpinnings of brain microvascular dysfunction in AD.
Collapse
Affiliation(s)
- Michelle A Erickson
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Harborview Medical Center, 325 9th Avenue, Seattle, WA, 98104, USA.
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA.
| | - Richard S Johnson
- Department of Genome Sciences, University of Washington, Seattle, USA
| | - Mamatha Damodarasamy
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Harborview Medical Center, 325 9th Avenue, Seattle, WA, 98104, USA
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, Seattle, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, Division of Neuropathology, University of Washington, Seattle, WA, USA
| | - William A Banks
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Harborview Medical Center, 325 9th Avenue, Seattle, WA, 98104, USA
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, 1660 S. Columbian Way, Seattle, WA, 98108, USA
| | - May J Reed
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Harborview Medical Center, 325 9th Avenue, Seattle, WA, 98104, USA.
| |
Collapse
|
3
|
Li X, Kempf S, Delgado Lagos F, Ukan Ü, Popp R, Hu J, Frömel T, Günther S, Weigert A, Fleming I. A regulatory loop involving the cytochrome P450-soluble epoxide hydrolase axis and TGF-β signaling. iScience 2024; 27:110938. [PMID: 39398242 PMCID: PMC11466655 DOI: 10.1016/j.isci.2024.110938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/11/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Fatty acid metabolites, produced by cytochrome P450 enzymes and soluble epoxide hydrolase (sEH), regulate inflammation. Here, we report that the transforming growth factor β (TGF-β)-induced polarization of macrophages to a pro-resolving phenotype requires Alk5 and Smad2 activation to increase sEH expression and activity. Macrophages lacking sEH showed impaired repolarization, reduced phagocytosis, and maintained a pro-inflammatory gene expression profile. 11,12-Epoxyeicosatrienoic acid (EET) was one altered metabolite in sEH-/- macrophages and mimicked the effect of sEH deletion on gene expression. Notably, 11,12-EET also reduced Alk5 expression, inhibiting TGF-β-induced Smad2 phosphorylation by triggering the cytosolic translocation of the E3 ligase Smurf2. These findings suggest that sEH expression is controlled by TGF-β and that sEH activity, which lowers 11,12-EET levels and promotes TGF-β signaling by metabolizing 11,12-EET to prevent Alk5 degradation. Thus, an autocrine loop between sEH/11,12-EET and TGF-β1 regulates macrophage function.
Collapse
Affiliation(s)
- Xiaoming Li
- Goethe University, Institute for Vascular Signalling, Centre for Molecular Medicine, Frankfurt am Main, Germany
| | - Sebastian Kempf
- Goethe University, Institute for Vascular Signalling, Centre for Molecular Medicine, Frankfurt am Main, Germany
| | - Fredy Delgado Lagos
- Goethe University, Institute for Vascular Signalling, Centre for Molecular Medicine, Frankfurt am Main, Germany
| | - Ürün Ukan
- Goethe University, Institute for Vascular Signalling, Centre for Molecular Medicine, Frankfurt am Main, Germany
| | - Rüdiger Popp
- Goethe University, Institute for Vascular Signalling, Centre for Molecular Medicine, Frankfurt am Main, Germany
| | - Jiong Hu
- Goethe University, Institute for Vascular Signalling, Centre for Molecular Medicine, Frankfurt am Main, Germany
- Department of Embryology and Histology, School of Basic Medicine, Tongi Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Timo Frömel
- Goethe University, Institute for Vascular Signalling, Centre for Molecular Medicine, Frankfurt am Main, Germany
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Andreas Weigert
- Goethe University, Institute of Biochemistry I, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Goethe University, Institute for Vascular Signalling, Centre for Molecular Medicine, Frankfurt am Main, Germany
- German Center of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| |
Collapse
|
4
|
Wojtas AM, Dammer EB, Guo Q, Ping L, Shantaraman A, Duong DM, Yin L, Fox EJ, Seifar F, Lee EB, Johnson ECB, Lah JJ, Levey AI, Levites Y, Rangaraju S, Golde TE, Seyfried NT. Proteomic changes in the human cerebrovasculature in Alzheimer's disease and related tauopathies linked to peripheral biomarkers in plasma and cerebrospinal fluid. Alzheimers Dement 2024; 20:4043-4065. [PMID: 38713744 PMCID: PMC11180878 DOI: 10.1002/alz.13821] [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: 01/25/2024] [Revised: 02/21/2024] [Accepted: 03/02/2024] [Indexed: 05/09/2024]
Abstract
INTRODUCTION Cerebrovascular dysfunction is a pathological hallmark of Alzheimer's disease (AD). Nevertheless, detecting cerebrovascular changes within bulk tissues has limited our ability to characterize proteomic alterations from less abundant cell types. METHODS We conducted quantitative proteomics on bulk brain tissues and isolated cerebrovasculature from the same individuals, encompassing control (N = 28), progressive supranuclear palsy (PSP) (N = 18), and AD (N = 21) cases. RESULTS Protein co-expression network analysis identified unique cerebrovascular modules significantly correlated with amyloid plaques, cerebrovascular amyloid angiopathy (CAA), and/or tau pathology. The protein products within AD genetic risk loci were concentrated within cerebrovascular modules. The overlap between differentially abundant proteins in AD cerebrospinal fluid (CSF) and plasma with cerebrovascular network highlighted a significant increase of matrisome proteins, SMOC1 and SMOC2, in CSF, plasma, and brain. DISCUSSION These findings enhance our understanding of cerebrovascular deficits in AD, shedding light on potential biomarkers associated with CAA and vascular dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Aleksandra M. Wojtas
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
| | - Eric B. Dammer
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
| | - Qi Guo
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
| | - Lingyan Ping
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
| | - Ananth Shantaraman
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
| | - Duc M. Duong
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
| | - Luming Yin
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
| | - Edward J. Fox
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
| | - Fatemeh Seifar
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
| | - Edward B. Lee
- Department of Pathology and Laboratory MedicineUniversity of PennsylvaniaPennsylvaniaUSA
| | - Erik C. B. Johnson
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
- Department of NeurologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - James J. Lah
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
- Department of NeurologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Allan I. Levey
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
- Department of NeurologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Yona Levites
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
- Department of Pharmacology and Chemical BiologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Srikant Rangaraju
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
- Department of NeurologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Todd E. Golde
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
- Department of NeurologyEmory University School of MedicineAtlantaGeorgiaUSA
- Department of Pharmacology and Chemical BiologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Nicholas T. Seyfried
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
- Center for Neurodegenerative DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
- Department of NeurologyEmory University School of MedicineAtlantaGeorgiaUSA
| |
Collapse
|
5
|
Aoki H, Takasawa A, Yamamoto E, Niinuma T, Yamano HO, Harada T, Kubo T, Yorozu A, Kitajima H, Ishiguro K, Kai M, Katanuma A, Shinohara T, Nakase H, Sugai T, Osanai M, Suzuki H. Downregulation of SMOC1 is associated with progression of colorectal traditional serrated adenomas. BMC Gastroenterol 2024; 24:91. [PMID: 38429655 PMCID: PMC10905814 DOI: 10.1186/s12876-024-03175-1] [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: 07/20/2023] [Accepted: 02/15/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Aberrant DNA methylation is prevalent in colorectal serrated lesions. We previously reported that the CpG island of SMOC1 is frequently methylated in traditional serrated adenomas (TSAs) and colorectal cancers (CRCs) but is rarely methylated in sessile serrated lesions (SSLs). In the present study, we aimed to further characterize the expression of SMOC1 in early colorectal lesions. METHODS SMOC1 expression was analyzed immunohistochemically in a series of colorectal tumors (n = 199) and adjacent normal colonic tissues (n = 112). RESULTS SMOC1 was abundantly expressed in normal colon and SSLs while it was significantly downregulated in TSAs, advanced adenomas and cancers. Mean immunohistochemistry scores were as follows: normal colon, 24.2; hyperplastic polyp (HP), 18.9; SSL, 23.8; SSL with dysplasia (SSLD)/SSL with early invasive cancer (EIC), 15.8; TSA, 5.4; TSA with high grade dysplasia (HGD)/EIC, 4.7; non-advanced adenoma, 21.4; advanced adenoma, 11.9; EIC, 10.9. Higher levels SMOC1 expression correlated positively with proximal colon locations and flat tumoral morphology, reflecting its abundant expression in SSLs. Among TSAs that contained both flat and protruding components, levels of SMOC1 expression were significantly lower in the protruding components. CONCLUSION Our results suggest that reduced expression of SMOC1 is associated with progression of TSAs and conventional adenomas and that SMOC1 expression may be a biomarker for diagnosis of serrated lesions and risk prediction in colorectal tumors.
Collapse
Affiliation(s)
- Hironori Aoki
- Department of Molecular Biology, Sapporo Medical University School of Medicine, S1, W17, Chuo-Ku, Sapporo, 060-8556, Japan
- Center for Gastroenterology, Teine-Keijinkai Hospital, Sapporo, Japan
- Department of Gastroenterology and Endoscopy, Koyukai Shin-Sapporo Hospital, Sapporo, Japan
| | - Akira Takasawa
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Eiichiro Yamamoto
- Department of Molecular Biology, Sapporo Medical University School of Medicine, S1, W17, Chuo-Ku, Sapporo, 060-8556, Japan
| | - Takeshi Niinuma
- Department of Molecular Biology, Sapporo Medical University School of Medicine, S1, W17, Chuo-Ku, Sapporo, 060-8556, Japan
| | - Hiro-O Yamano
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Taku Harada
- Department of Molecular Biology, Sapporo Medical University School of Medicine, S1, W17, Chuo-Ku, Sapporo, 060-8556, Japan
- Center for Gastroenterology, Teine-Keijinkai Hospital, Sapporo, Japan
| | - Toshiyuki Kubo
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Akira Yorozu
- Department of Molecular Biology, Sapporo Medical University School of Medicine, S1, W17, Chuo-Ku, Sapporo, 060-8556, Japan
| | - Hiroshi Kitajima
- Department of Molecular Biology, Sapporo Medical University School of Medicine, S1, W17, Chuo-Ku, Sapporo, 060-8556, Japan
| | - Kazuya Ishiguro
- Department of Molecular Biology, Sapporo Medical University School of Medicine, S1, W17, Chuo-Ku, Sapporo, 060-8556, Japan
| | - Masahiro Kai
- Department of Molecular Biology, Sapporo Medical University School of Medicine, S1, W17, Chuo-Ku, Sapporo, 060-8556, Japan
| | - Akio Katanuma
- Center for Gastroenterology, Teine-Keijinkai Hospital, Sapporo, Japan
| | | | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tamotsu Sugai
- Department of Molecular Diagnostic Pathology, Iwate Medical University, Morioka, Japan
| | - Makoto Osanai
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiromu Suzuki
- Department of Molecular Biology, Sapporo Medical University School of Medicine, S1, W17, Chuo-Ku, Sapporo, 060-8556, Japan.
| |
Collapse
|
6
|
Ahuja S, Zaheer S. Multifaceted TGF-β signaling, a master regulator: From bench-to-bedside, intricacies, and complexities. Cell Biol Int 2024; 48:87-127. [PMID: 37859532 DOI: 10.1002/cbin.12097] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Physiological embryogenesis and adult tissue homeostasis are regulated by transforming growth factor-β (TGF-β), an evolutionarily conserved family of secreted polypeptide factors, acting in an autocrine and paracrine manner. The role of TGF-β in inflammation, fibrosis, and cancer is complex and sometimes even contradictory, exhibiting either inhibitory or promoting effects depending on the stage of the disease. Under pathological conditions, especially fibrosis and cancer, overexpressed TGF-β causes extracellular matrix deposition, epithelial-mesenchymal transition, cancer-associated fibroblast formation, and/or angiogenesis. In this review article, we have tried to dive deep into the mechanism of action of TGF-β in inflammation, fibrosis, and carcinogenesis. As TGF-β and its downstream signaling mechanism are implicated in fibrosis and carcinogenesis blocking this signaling mechanism appears to be a promising avenue. However, targeting TGF-β carries substantial risk as this pathway is implicated in multiple homeostatic processes and is also known to have tumor-suppressor functions. There is a need for careful dosing of TGF-β drugs for therapeutic use and patient selection.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
7
|
Wojtas AM, Dammer EB, Guo Q, Ping L, Shantaraman A, Duong DM, Yin L, Fox EJ, Seifar F, Lee EB, Johnson ECB, Lah JJ, Levey AI, Levites Y, Rangaraju S, Golde TE, Seyfried NT. Proteomic Changes in the Human Cerebrovasculature in Alzheimer's Disease and Related Tauopathies Linked to Peripheral Biomarkers in Plasma and Cerebrospinal Fluid. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.10.24301099. [PMID: 38260316 PMCID: PMC10802758 DOI: 10.1101/2024.01.10.24301099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Dysfunction of the neurovascular unit stands as a significant pathological hallmark of Alzheimer's disease (AD) and age-related neurodegenerative diseases. Nevertheless, detecting vascular changes in the brain within bulk tissues has proven challenging, limiting our ability to characterize proteomic alterations from less abundant cell types. To address this challenge, we conducted quantitative proteomic analyses on both bulk brain tissues and cerebrovascular-enriched fractions from the same individuals, encompassing cognitively unimpaired control, progressive supranuclear palsy (PSP), and AD cases. Protein co-expression network analysis identified modules unique to the cerebrovascular fractions, specifically enriched with pericytes, endothelial cells, and smooth muscle cells. Many of these modules also exhibited significant correlations with amyloid plaques, cerebral amyloid angiopathy (CAA), and/or tau pathology in the brain. Notably, the protein products within AD genetic risk loci were found concentrated within modules unique to the vascular fractions, consistent with a role of cerebrovascular deficits in the etiology of AD. To prioritize peripheral AD biomarkers associated with vascular dysfunction, we assessed the overlap between differentially abundant proteins in AD cerebrospinal fluid (CSF) and plasma with a vascular-enriched network modules in the brain. This analysis highlighted matrisome proteins, SMOC1 and SMOC2, as being increased in CSF, plasma, and brain. Immunohistochemical analysis revealed SMOC1 deposition in both parenchymal plaques and CAA in the AD brain, whereas SMOC2 was predominantly localized to CAA. Collectively, these findings significantly enhance our understanding of the involvement of cerebrovascular abnormalities in AD, shedding light on potential biomarkers and molecular pathways associated with CAA and vascular dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Aleksandra M. Wojtas
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric B. Dammer
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Qi Guo
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Lingyan Ping
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Ananth Shantaraman
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Duc M. Duong
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Luming Yin
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Edward J. Fox
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Fatemeh Seifar
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Edward B. Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, PA, USA
| | - Erik C. B. Johnson
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - James J. Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Allan I. Levey
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Yona Levites
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Srikant Rangaraju
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Todd E. Golde
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Nicholas T. Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
8
|
Li J, Yao YX, Yao PS. Circulating Biomarkers and Risk of Hypertension: A Two-Sample Mendelian Randomisation Study. Heart Lung Circ 2023; 32:1434-1442. [PMID: 38042639 DOI: 10.1016/j.hlc.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/27/2023] [Accepted: 09/02/2023] [Indexed: 12/04/2023]
Abstract
OBJECTIVE This study systematically assessed circulating proteins to identify new serum biomarkers and risk of hypertension using Mendelian randomisation. METHODS The associations between 4,782 human circulating proteins and the risk of hypertension were evaluated using two-sample Mendelian randomisation. The FinnGen study demonstrated a link between genetic predisposition and hypertension in 85,438 cases and 223,663 controls. RESULTS Inverse variance weighted and sensitivity analysis revealed nine proteins in circulation that have a causative effect on hypertension. SMOC1 and TIE1 were determined to be causative factors in the decreased likelihood of developing hypertension, with odds ratios of 0.86 (95% CI 0.81-0.91; p=1.06e-06) and 0.96 (95% CI 0.94-0.98; p=9.39e-05), respectively. NDUFB4, ETHE1, POFUT2, TRIL, ADAM23, GXYLT1, OXT, TPST2, and TMCC3 showed a possible connection to hypertension. CONCLUSIONS This two-sample Mendelian randomisation study found that SMOC1 and TIE1 are causally linked to hypertension, making them a promising target for therapy.
Collapse
Affiliation(s)
- Jin Li
- Department of Cardiovascular Medicine, Fujian Provincial Geriatric Hospital, Fuzhou, China
| | - Yue-Xian Yao
- Department of Cardiovascular Medicine, Fujian Provincial Geriatric Hospital, Fuzhou, China
| | - Pei-Sen Yao
- Department of Neurosurgery, Neurosurgical Research Institute, First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
9
|
Kwon W, Yoo C, Kim JH, Kim T, Kim A, Hwang M, Choi H. Role of human dural fibroblasts in the angiogenic responses of human endothelial cells: An in vitro dural model and the application of lab-on-a-chip for EDAS. Bioeng Transl Med 2023; 8:e10589. [PMID: 38023706 PMCID: PMC10658529 DOI: 10.1002/btm2.10589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/18/2023] [Accepted: 08/01/2023] [Indexed: 12/01/2023] Open
Abstract
Encephaloduroarteriosynangiosis (EDAS), an indirect anastomosis procedure, is widely accepted as a primary treatment for moyamoya disease (MMD) to improve collateral blood flow. During surgical intervention, dural fibroblasts (DuF) are thought to produce various proteins that create an angiogenic microenvironment. However, the biophysiological evidence supporting the angiogenic properties of this surgical technique has not been thoroughly elucidated. The purpose of these studies was to determine whether DuF releases pro-angiogenic factors and chemokines and promotes angiogenic properties in human endothelial cells (ECs) under IL-1β-mediated wound conditions, which are expected to occur during the process of neo-vascularization within the dura mater. Furthermore, a microfluidic chemotaxis platform was implemented to investigate the angiogenic activity of ECs in response to a reconstituted dura model. Transcriptome sequencing revealed that IL-1β stimulation on DuF induced a significant upregulation of various pro-angiogenic genes, including IL-6, IL-8, CCL-2, CCL-5, SMOC-1, and SCG-2 (p < 0.05). Moreover, compared to ECs cultured in naïve media or naïve DuF media, those exposed to IL-1β-DuF conditioned media expressed higher mRNA and protein levels of these pro-angiogenic factors (p < 0.001). ECs co-cultured with IL-1β-DuF also exhibited considerable migration on the microfluidic chemotaxis platform. Furthermore, the chemotactic effects on the ECs were reduced upon neutralization of IL-8 or inhibition of NF-κB signaling. Our findings demonstrate that IL-1β-DuFs release factors that activate and enhance the angiogenic properties of ECs. These results suggest a potential interaction between DuF and ECs following EDAS for MMD, and these components could be targeted for the development of therapeutic biomarkers.
Collapse
Affiliation(s)
- Woo‐Keun Kwon
- Department of Neurosurgery, Korea University Guro HospitalKorea University College of MedicineSeoulSouth Korea
| | - Chang‐Min Yoo
- Department of Medical Sciences, Graduate School of MedicineKorea UniversitySeoulSouth Korea
| | - Jang Hun Kim
- Department of Neurosurgery, Korea University Anam HospitalKorea University College of MedicineSeoulSouth Korea
| | - Tae‐Won Kim
- Department of Medical Sciences, Graduate School of MedicineKorea UniversitySeoulSouth Korea
| | - An‐Gi Kim
- Department of Medical Sciences, Graduate School of MedicineKorea UniversitySeoulSouth Korea
| | - Min‐Ho Hwang
- Department of Medical Sciences, Graduate School of MedicineKorea UniversitySeoulSouth Korea
| | - Hyuk Choi
- Department of Medical Sciences, Graduate School of MedicineKorea UniversitySeoulSouth Korea
| |
Collapse
|
10
|
Roberts JA, Varma VR, Candia J, Tanaka T, Ferrucci L, Bennett DA, Thambisetty M. Unbiased proteomics and multivariable regularized regression techniques identify SMOC1, NOG, APCS, and NTN1 in an Alzheimer's disease brain proteomic signature. NPJ AGING 2023; 9:18. [PMID: 37414805 PMCID: PMC10326005 DOI: 10.1038/s41514-023-00112-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 05/18/2023] [Indexed: 07/08/2023]
Abstract
Advancements in omics methodologies have generated a wealth of high-dimensional Alzheimer's disease (AD) datasets, creating significant opportunities and challenges for data interpretation. In this study, we utilized multivariable regularized regression techniques to identify a reduced set of proteins that could discriminate between AD and cognitively normal (CN) brain samples. Utilizing eNetXplorer, an R package that tests the accuracy and significance of a family of elastic net generalized linear models, we identified 4 proteins (SMOC1, NOG, APCS, NTN1) that accurately discriminated between AD (n = 31) and CN (n = 22) middle frontal gyrus (MFG) tissue samples from Religious Orders Study participants with 83 percent accuracy. We then validated this signature in MFG samples from Baltimore Longitudinal Study of Aging participants using leave-one-out logistic regression cross-validation, finding that the signature again accurately discriminated AD (n = 31) and CN (n = 19) participants with a receiver operating characteristic curve area under the curve of 0.863. These proteins were strongly correlated with the burden of neurofibrillary tangle and amyloid pathology in both study cohorts. We additionally tested whether these proteins differed between AD and CN inferior temporal gyrus (ITG) samples and blood serum samples at the time of AD diagnosis in ROS and BLSA, finding that the proteins differed between AD and CN ITG samples but not in blood serum samples. The identified proteins may provide mechanistic insights into the pathophysiology of AD, and the methods utilized in this study may serve as the basis for further work with additional high-dimensional datasets in AD.
Collapse
Affiliation(s)
- Jackson A Roberts
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA.
| | - Vijay R Varma
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Julián Candia
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Toshiko Tanaka
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
11
|
Tavallaee G, Lively S, Rockel JS, Ali SA, Im M, Sarda C, Mitchell GM, Rossomacha E, Nakamura S, Potla P, Gabrial S, Matelski J, Ratneswaran A, Perry K, Hinz B, Gandhi R, Jurisica I, Kapoor M. Contribution of MicroRNA-27b-3p to Synovial Fibrotic Responses in Knee Osteoarthritis. Arthritis Rheumatol 2022; 74:1928-1942. [PMID: 35791923 PMCID: PMC10946865 DOI: 10.1002/art.42285] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 05/18/2022] [Accepted: 06/23/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Synovial fibrosis contributes to osteoarthritis (OA) pathology, but the underlying mechanisms remain unknown. We have observed increased microRNA-27b-3p (miR-27b-3p) levels in synovial fluid of patients with late-stage radiographic knee OA. Here, we investigated the contribution of miR-27b-3p to synovial fibrosis in patients with severe knee OA and in a mouse model of knee OA. METHODS We stained synovium sections obtained from patients with radiographic knee OA scored according to the Kellgren/Lawrence scale and mice that underwent destabilization of the medial meniscus (DMM) for miR-27b-3p using in situ hybridization. We examined the effects of intraarticular injection of miR-27b-3p mimic into naive mouse knee joints and intraarticular injection of a miR-27b-3p inhibitor into mouse knee joints after DMM. We performed transfection with miR-27b-3p mimic and miR-27b-3p inhibitor in human OA fibroblast-like synoviocytes (FLS) using reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) array, RNA sequencing, RT-qPCR, Western blotting, immunofluorescence, and migration assays. RESULTS We observed increased miR-27b-3p expression in the synovium from patients with knee OA and in mice with DMM-induced arthritis. Injection of the miR-27b-3p mimic in mouse knee joints induced a synovial fibrosis-like phenotype, increased synovitis scores, and increased COL1A1 and α-smooth muscle actin (α-SMA) expression. In the mouse model of DMM-induced arthritis, injection of the miR-27b-3p inhibitor decreased α-SMA but did not change COL1A1 expression levels or synovitis scores. Transfection with the miR-27b-3p mimic in human OA FLS induced profibrotic responses, including increased migration and expression of key extracellular matrix (ECM) genes, but transfection with the miR-27b-3p inhibitor had the opposite effects. RNA sequencing identified a PPARG/ADAMTS8 signaling axis regulated by miR-27b-3p in OA FLS. Human OA FLS transfected with miR-27b-3p mimic and then treated with the PPARG agonist rosiglitazone or with ADAMTS8 small interfering RNA exhibited altered expression of select ECM genes. CONCLUSION Our findings demonstrate that miR-27b-3p has a key role in ECM regulation associated with synovial fibrosis during OA.
Collapse
Affiliation(s)
- Ghazaleh Tavallaee
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Krembil Research Institute, University Health Network, and Department of Laboratory Medicine and Pathobiology, University of TorontoTorontoOntarioCanada
| | - Starlee Lively
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, and Krembil Research Institute, University Health NetworkTorontoOntarioCanada
| | - Jason S. Rockel
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, and Krembil Research Institute, University Health NetworkTorontoOntarioCanada
| | - Shabana Amanda Ali
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada, and Bone & Joint Center, Department of Orthopaedic Surgery, Henry Ford Health SystemDetroitMichigan
| | - Michelle Im
- Faculty of Dentistry, University of TorontoTorontoOntarioCanada
| | - Clementine Sarda
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, and Krembil Research Institute, University Health NetworkTorontoOntarioCanada
| | - Greniqueca M. Mitchell
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, and Krembil Research Institute, University Health NetworkTorontoOntarioCanada
| | - Evgeny Rossomacha
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, and Krembil Research Institute, University Health NetworkTorontoOntarioCanada
| | - Sayaka Nakamura
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, and Krembil Research Institute, University Health NetworkTorontoOntarioCanada
| | - Pratibha Potla
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, and Krembil Research Institute, University Health NetworkTorontoOntarioCanada
| | - Sarah Gabrial
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, and Krembil Research Institute, University Health NetworkTorontoOntarioCanada
| | - John Matelski
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, and Krembil Research Institute, University Health NetworkTorontoOntarioCanada
| | - Anusha Ratneswaran
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, and Krembil Research Institute, University Health NetworkTorontoOntarioCanada
| | - Kim Perry
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, and Krembil Research Institute, University Health NetworkTorontoOntarioCanada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, and Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael's HospitalTorontoOntarioCanada
| | - Rajiv Gandhi
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Krembil Research Institute, University Health Network, and Departments of Medical Biophysics and Computer Science, University of TorontoTorontoOntarioCanada
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Krembil Research Institute, University Health Network, Toronto, Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, Ontario, Canada, and Institute of Neuroimmunology, Slovak Academy of SciencesBratislavaSlovakia
| | - Mohit Kapoor
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Krembil Research Institute, University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, and Division of Orthopaedic Surgery, Department of Surgery, University of TorontoTorontoOntarioCanada
| |
Collapse
|
12
|
Gul H, Habib G, Khan IM, Rahman SU, Khan NM, Wang H, Khan NU, Liu Y. Genetic resilience in chickens against bacterial, viral and protozoal pathogens. Front Vet Sci 2022; 9:1032983. [PMID: 36439341 PMCID: PMC9691405 DOI: 10.3389/fvets.2022.1032983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/28/2022] [Indexed: 06/13/2024] Open
Abstract
The genome contributes to the uniqueness of an individual breed, and enables distinctive characteristics to be passed from one generation to the next. The allelic heterogeneity of a certain breed results in a different response to a pathogen with different genomic expression. Disease resistance in chicken is a polygenic trait that involves different genes that confer resistance against pathogens. Such resistance also involves major histocompatibility (MHC) molecules, immunoglobulins, cytokines, interleukins, T and B cells, and CD4+ and CD8+ T lymphocytes, which are involved in host protection. The MHC is associated with antigen presentation, antibody production, and cytokine stimulation, which highlight its role in disease resistance. The natural resistance-associated macrophage protein 1 (Nramp-1), interferon (IFN), myxovirus-resistance gene, myeloid differentiation primary response 88 (MyD88), receptor-interacting serine/threonine kinase 2 (RIP2), and heterophile cells are involved in disease resistance and susceptibility of chicken. Studies related to disease resistance genetics, epigenetics, and quantitative trait loci would enable the identification of resistance markers and the development of disease resistance breeds. Microbial infections are responsible for significant outbreaks and have blighted the poultry industry. Breeding disease-resistant chicken strains may be helpful in tackling pathogens and increasing the current understanding on host genetics in the fight against communicable diseases. Advanced technologies, such as the CRISPR/Cas9 system, whole genome sequencing, RNA sequencing, and high-density single nucleotide polymorphism (SNP) genotyping, aid the development of resistant breeds, which would significantly decrease the use of antibiotics and vaccination in poultry. In this review, we aimed to reveal the recent genetic basis of infection and genomic modification that increase resistance against different pathogens in chickens.
Collapse
Affiliation(s)
- Haji Gul
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Gul Habib
- Department of Microbiology, Abbottabad University of Science and Technology, Abbottabad, Pakistan
| | - Ibrar Muhammad Khan
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Sajid Ur Rahman
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Nazir Muhammad Khan
- Department of Zoology, University of Science and Technology, Bannu, Pakistan
| | - Hongcheng Wang
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Najeeb Ullah Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, Pakistan
| | - Yong Liu
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| |
Collapse
|
13
|
Gauer JS, Ajjan RA, Ariëns RAS. Platelet-Neutrophil Interaction and Thromboinflammation in Diabetes: Considerations for Novel Therapeutic Approaches. J Am Heart Assoc 2022; 11:e027071. [PMID: 36250653 DOI: 10.1161/jaha.122.027071] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Thromboinflammation has become a topic of key interest in cardiovascular disease and the prevention of diabetes complications because of the interplay between thrombosis and inflammation in diabetes. Specifically, the significant risk of vascular thrombotic disease in diabetes highlights the need for new and better therapeutic targets to help manage and prevent vascular thrombo-occlusive disease in this condition. Similarly, the prominent role of inflammation in diabetes has sparked interest in anti-inflammatory agents to better prevent and control vascular disease. Investigations on the effects of anticoagulation and antiplatelet interventions in patients with diabetes and cardiovascular disease show a potential role for these agents in decreasing morbidity and mortality. Neutrophils and platelets are key players in inflammation and wound-healing response, respectively. The interaction between neutrophils and platelets is thought to be an important driver of thromboinflammation. Therefore, this review describes the mechanisms involved in platelet-neutrophil interactions that contribute to the development or exacerbation of thromboinflammation in the context of diabetes and its associated comorbidities. The effects observed by the antithrombotic/antidiabetic treatments and physical activity/dietary interventions on attenuating thromboinflammation are discussed. These data suggest that mechanisms involved in platelet-neutrophil interaction, platelet activation/aggregation, and the recruitment of neutrophils have a promising potential to become therapeutic targets to decrease thromboinflammation in patients with diabetes.
Collapse
Affiliation(s)
- Julia S Gauer
- Discovery and Translational Science Department Institute of Cardiovascular and Metabolic Medicine, University of Leeds Leeds United Kingdom
| | - Ramzi A Ajjan
- Discovery and Translational Science Department Institute of Cardiovascular and Metabolic Medicine, University of Leeds Leeds United Kingdom
| | - Robert A S Ariëns
- Discovery and Translational Science Department Institute of Cardiovascular and Metabolic Medicine, University of Leeds Leeds United Kingdom
| |
Collapse
|
14
|
Recouvreux MS, Miao J, Gozo MC, Wu J, Walts AE, Karlan BY, Orsulic S. FOXC2 Promotes Vasculogenic Mimicry in Ovarian Cancer. Cancers (Basel) 2022; 14:4851. [PMID: 36230774 PMCID: PMC9564305 DOI: 10.3390/cancers14194851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
FOXC2 is a forkhead family transcription factor that plays a critical role in specifying mesenchymal cell fate during embryogenesis. FOXC2 expression is associated with increased metastasis and poor survival in various solid malignancies. Using in vitro and in vivo assays in mouse ovarian cancer cell lines, we confirmed the previously reported mechanisms by which FOXC2 could promote cancer growth, metastasis, and drug resistance, including epithelial-mesenchymal transition, stem cell-like differentiation, and resistance to anoikis. In addition, we showed that FOXC2 expression is associated with vasculogenic mimicry in mouse and human ovarian cancers. FOXC2 overexpression increased the ability of human ovarian cancer cells to form vascular-like structures in vitro, while inhibition of FOXC2 had the opposite effect. Thus, we present a novel mechanism by which FOXC2 might contribute to cancer aggressiveness and poor patient survival.
Collapse
Affiliation(s)
- Maria Sol Recouvreux
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jiangyong Miao
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Maricel C. Gozo
- Women’s Cancer Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jingni Wu
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sandra Orsulic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90095, USA
| |
Collapse
|
15
|
Hachana S, Larrivée B. TGF-β Superfamily Signaling in the Eye: Implications for Ocular Pathologies. Cells 2022; 11:2336. [PMID: 35954181 PMCID: PMC9367584 DOI: 10.3390/cells11152336] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/06/2023] Open
Abstract
The TGF-β signaling pathway plays a crucial role in several key aspects of development and tissue homeostasis. TGF-β ligands and their mediators have been shown to be important regulators of ocular physiology and their dysregulation has been described in several eye pathologies. TGF-β signaling participates in regulating several key developmental processes in the eye, including angiogenesis and neurogenesis. Inadequate TGF-β signaling has been associated with defective angiogenesis, vascular barrier function, unfavorable inflammatory responses, and tissue fibrosis. In addition, experimental models of corneal neovascularization, diabetic retinopathy, proliferative vitreoretinopathy, glaucoma, or corneal injury suggest that aberrant TGF-β signaling may contribute to the pathological features of these conditions, showing the potential of modulating TGF-β signaling to treat eye diseases. This review highlights the key roles of TGF-β family members in ocular physiology and in eye diseases, and reviews approaches targeting the TGF-β signaling as potential treatment options.
Collapse
Affiliation(s)
- Soumaya Hachana
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Bruno Larrivée
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| |
Collapse
|
16
|
Proteomic profiling of concurrently isolated primary microvascular endothelial cells, pericytes, and vascular smooth muscle cells from adult mouse heart. Sci Rep 2022; 12:8835. [PMID: 35614104 PMCID: PMC9132906 DOI: 10.1038/s41598-022-12749-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
The microcirculation serves crucial functions in adult heart, distinct from those carried out by epicardial vessels. Microvessels are governed by unique regulatory mechanisms, impairment of which leads to microvessel-specific pathology. There are few treatment options for patients with microvascular heart disease, primarily due to limited understanding of underlying pathology. High throughput mRNA sequencing and protein expression profiling in specific cells can improve our understanding of microvessel biology and disease at the molecular level. Understanding responses of individual microvascular cells to the same physiological or pathophysiological stimuli requires the ability to isolate the specific cell types that comprise the functional units of the microcirculation in the heart, preferably from the same heart, to ensure that different cells have been exposed to the same in-vivo conditions. We developed an integrated process for simultaneous isolation and culture of the main cell types comprising the microcirculation in adult mouse heart: endothelial cells, pericytes, and vascular smooth muscle cells. These cell types were characterized with isobaric labeling quantitative proteomics and mRNA sequencing. We defined microvascular cell proteomes, identified novel protein markers, and confirmed established cell-specific markers. Our results allow identification of unique markers and regulatory proteins that govern microvascular physiology and pathology.
Collapse
|
17
|
Ukan Ü, Delgado Lagos F, Kempf S, Günther S, Siragusa M, Fisslthaler B, Fleming I. Effect of Thrombin on the Metabolism and Function of Murine Macrophages. Cells 2022; 11:cells11101718. [PMID: 35626753 PMCID: PMC9139186 DOI: 10.3390/cells11101718] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Macrophages are plastic and heterogeneous immune cells that adapt pro- or anti-inflammatory phenotypes upon exposure to different stimuli. Even though there has been evidence supporting a crosstalk between coagulation and innate immunity, the way in which protein components of the hemostasis pathway influence macrophages remains unclear. We investigated the effect of thrombin on macrophage polarization. On the basis of gene expression and cytokine secretion, our results suggest that polarization with thrombin induces an anti-inflammatory, M2-like phenotype. In functional studies, thrombin polarization promoted oxLDL phagocytosis by macrophages, and conditioned medium from the same cells increased endothelial cell proliferation. There were, however, clear differences between the classical M2a polarization and the effects of thrombin on gene expression. Finally, the deletion and inactivation of secreted modular Ca2+-binding protein 1 (SMOC1) attenuated phagocytosis by thrombin-stimulated macrophages, a phenomenon revered by the addition of recombinant SMOC1. Manipulation of SMOC1 levels also had a pronounced impact on the expression of TGF-β-signaling-related genes. Taken together, our results show that thrombin induces an anti-inflammatory macrophage phenotype with similarities as well as differences to the classical alternatively activated M2 polarization states, highlighting the importance of tissue levels of SMOC1 in modifying thrombin-induced macrophage polarization.
Collapse
Affiliation(s)
- Ürün Ukan
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany; (Ü.U.); (F.D.L.); (S.K.); (M.S.); (B.F.)
| | - Fredy Delgado Lagos
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany; (Ü.U.); (F.D.L.); (S.K.); (M.S.); (B.F.)
| | - Sebastian Kempf
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany; (Ü.U.); (F.D.L.); (S.K.); (M.S.); (B.F.)
| | - Stefan Günther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany;
| | - Mauro Siragusa
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany; (Ü.U.); (F.D.L.); (S.K.); (M.S.); (B.F.)
| | - Beate Fisslthaler
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany; (Ü.U.); (F.D.L.); (S.K.); (M.S.); (B.F.)
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany; (Ü.U.); (F.D.L.); (S.K.); (M.S.); (B.F.)
- German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, 60596 Frankfurt am Main, Germany
- CardioPulmonary Institute, Goethe University, 60596 Frankfurt am Main, Germany
- Correspondence:
| |
Collapse
|
18
|
Drummond E, Kavanagh T, Pires G, Marta-Ariza M, Kanshin E, Nayak S, Faustin A, Berdah V, Ueberheide B, Wisniewski T. The amyloid plaque proteome in early onset Alzheimer's disease and Down syndrome. Acta Neuropathol Commun 2022; 10:53. [PMID: 35418158 PMCID: PMC9008934 DOI: 10.1186/s40478-022-01356-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/28/2022] [Indexed: 11/17/2022] Open
Abstract
Amyloid plaques contain many proteins in addition to beta amyloid (Aβ). Previous studies examining plaque-associated proteins have shown these additional proteins are important; they provide insight into the factors that drive amyloid plaque development and are potential biomarkers or therapeutic targets for Alzheimer's disease (AD). The aim of this study was to comprehensively identify proteins that are enriched in amyloid plaques using unbiased proteomics in two subtypes of early onset AD: sporadic early onset AD (EOAD) and Down Syndrome (DS) with AD. We focused our study on early onset AD as the drivers of the more aggressive pathology development in these cases is unknown and it is unclear whether amyloid-plaque enriched proteins differ between subtypes of early onset AD. Amyloid plaques and neighbouring non-plaque tissue were microdissected from human brain sections using laser capture microdissection and label-free LC-MS was used to quantify the proteins present. 48 proteins were consistently enriched in amyloid plaques in EOAD and DS. Many of these proteins were more significantly enriched in amyloid plaques than Aβ. The most enriched proteins in amyloid plaques in both EOAD and DS were: COL25A1, SMOC1, MDK, NTN1, OLFML3 and HTRA1. Endosomal/lysosomal proteins were particularly highly enriched in amyloid plaques. Fluorescent immunohistochemistry was used to validate the enrichment of four proteins in amyloid plaques (moesin, ezrin, ARL8B and SMOC1) and to compare the amount of total Aβ, Aβ40, Aβ42, phosphorylated Aβ, pyroglutamate Aβ species and oligomeric species in EOAD and DS. These studies showed that phosphorylated Aβ, pyroglutamate Aβ species and SMOC1 were significantly higher in DS plaques, while oligomers were significantly higher in EOAD. Overall, we observed that amyloid plaques in EOAD and DS largely contained the same proteins, however the amount of enrichment of some proteins was different in EOAD and DS. Our study highlights the significant enrichment of many proteins in amyloid plaques, many of which may be potential therapeutic targets and/or biomarkers for AD.
Collapse
Affiliation(s)
- Eleanor Drummond
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia.
- Centre for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, Science Building, Rm 1017, 435 East 30th Street, New York, NY, 10016, USA.
| | - Tomas Kavanagh
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Geoffrey Pires
- Centre for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, Science Building, Rm 1017, 435 East 30th Street, New York, NY, 10016, USA
| | - Mitchell Marta-Ariza
- Centre for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, Science Building, Rm 1017, 435 East 30th Street, New York, NY, 10016, USA
| | - Evgeny Kanshin
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, USA
| | - Shruti Nayak
- Merck & Co., Inc, Computational & Structural Chemistry, Kenilworth, NJ, USA
| | - Arline Faustin
- Centre for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, Science Building, Rm 1017, 435 East 30th Street, New York, NY, 10016, USA
| | - Valentin Berdah
- Centre for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, Science Building, Rm 1017, 435 East 30th Street, New York, NY, 10016, USA
| | - Beatrix Ueberheide
- Centre for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, Science Building, Rm 1017, 435 East 30th Street, New York, NY, 10016, USA
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Thomas Wisniewski
- Centre for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, Science Building, Rm 1017, 435 East 30th Street, New York, NY, 10016, USA.
- Departments of Pathology and Psychiatry, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
19
|
Sekulic-Jablanovic M, Paproth J, Sgambato C, Albano G, Fuster DG, Bodmer D, Petkovic V. Lack of NHE6 and Inhibition of NKCC1 Associated With Increased Permeability in Blood Labyrinth Barrier-Derived Endothelial Cell Layer. Front Cell Neurosci 2022; 16:862119. [PMID: 35496913 PMCID: PMC9039518 DOI: 10.3389/fncel.2022.862119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/17/2022] [Indexed: 12/20/2022] Open
Abstract
Acoustic trauma, autoimmune inner ear disease, and presbycusis feature loss of the integrity of the blood-labyrinth barrier (BLB). Normal BLB function depends on endothelial structural integrity, which is supported and maintained by tight junctions and adherens junctions within the microvascular endothelial layer. When these junctions are disrupted, vascular leakage occurs. Tight junctions and adherens junctions are functionally and structurally linked, but the exact signaling pathways underlying their interaction remain unknown. In addition, solute carriers (SC) are essential for optimal exchange through BLB. Previously, we found that SC family member, the sodium–hydrogen exchanger NHE6, was expressed in all wildtype cochlear tissues, and that Nhe6-knockout mice displayed moderate hearing loss. Moreover, NHE6 depletion affected Trk protein turnover and endosomal signaling. Here, we investigated whether NHE6 might impact BLB integrity. We found that Nhe6-knockout, BLB-derived endothelial cells showed reduced expression of major junctional genes: Tjp1, F11r, Ocln, Cdh5, and Cldn5. Co-culturing BLB-derived endothelial cells with pericytes and/or perivascular resident macrophage-like melanocytes in a transwell system showed that monolayers of Nhe6-knockout BLB-derived cells had lower electrical resistance and higher permeability, compared to wildtype endothelial monolayers. Additionally, another SC, NKCC1, which was previously linked to congenital deafness, was downregulated in our Nhe6-knockout mouse model. Blocking NKCC1 with a NKCC1-specific inhibitor, bumetanide, in wildtype BLB-derived endothelial cells also caused the downregulation of major junctional proteins, particularly Tjp1 and F11r, which encode the zonula occludens and junctional adhesion molecule-1 proteins, respectively. Moreover, bumetanide treatment increased cell permeability. In conclusion, we showed that the lack or inhibition of NHE6 or NKCC1 affected the permeability of endothelial BLB-derived cells. These findings suggested that NHE6 and NKCC1 could serve as potential targets for modifying BLB permeability to facilitate drug delivery across the BLB to the cochlea or to protect the cochlea from ototoxic insults.
Collapse
Affiliation(s)
- Marijana Sekulic-Jablanovic
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- *Correspondence: Marijana Sekulic-Jablanovic,
| | - Jessica Paproth
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Cinzia Sgambato
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Giuseppe Albano
- Inselspital Bern, Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Daniel G. Fuster
- Inselspital Bern, Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Daniel Bodmer
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Clinic for Otolaryngology, Head and Neck Surgery, University Hospital Basel, Basel, Switzerland
| | - Vesna Petkovic
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
20
|
Kim TH, Hong DG, Yang YM. Hepatokines and Non-Alcoholic Fatty Liver Disease: Linking Liver Pathophysiology to Metabolism. Biomedicines 2021; 9:biomedicines9121903. [PMID: 34944728 PMCID: PMC8698516 DOI: 10.3390/biomedicines9121903] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/12/2021] [Accepted: 12/12/2021] [Indexed: 12/16/2022] Open
Abstract
The liver plays a key role in maintaining energy homeostasis by sensing and responding to changes in nutrient status under various metabolic conditions. Recently highlighted as a major endocrine organ, the contribution of the liver to systemic glucose and lipid metabolism is primarily attributed to signaling crosstalk between multiple organs via hepatic hormones, cytokines, and hepatokines. Hepatokines are hormone-like proteins secreted by hepatocytes, and a number of these have been associated with extra-hepatic metabolic regulation. Mounting evidence has revealed that the secretory profiles of hepatokines are significantly altered in non-alcoholic fatty liver disease (NAFLD), the most common hepatic manifestation, which frequently precedes other metabolic disorders, including insulin resistance and type 2 diabetes. Therefore, deciphering the mechanism of hepatokine-mediated inter-organ communication is essential for understanding the complex metabolic network between tissues, as well as for the identification of novel diagnostic and/or therapeutic targets in metabolic disease. In this review, we describe the hepatokine-driven inter-organ crosstalk in the context of liver pathophysiology, with a particular focus on NAFLD progression. Moreover, we summarize key hepatokines and their molecular mechanisms of metabolic control in non-hepatic tissues, discussing their potential as novel biomarkers and therapeutic targets in the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Tae Hyun Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women’s University, Seoul 04310, Korea;
| | - Dong-Gyun Hong
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Korea;
- KNU Researcher Training Program for Developing Anti-Viral Innovative Drugs, Kangwon National University, Chuncheon 24341, Korea
| | - Yoon Mee Yang
- Department of Pharmacy, Kangwon National University, Chuncheon 24341, Korea;
- KNU Researcher Training Program for Developing Anti-Viral Innovative Drugs, Kangwon National University, Chuncheon 24341, Korea
- Correspondence: ; Tel.: +82-33-250-6909
| |
Collapse
|
21
|
Wang J, Xia S, Zhao J, Gong C, Xi Q, Sun W. Prognostic Potential of Secreted Modular Calcium-Binding Protein 1 in Low-Grade Glioma. Front Mol Biosci 2021; 8:666623. [PMID: 34869577 PMCID: PMC8640086 DOI: 10.3389/fmolb.2021.666623] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 10/20/2021] [Indexed: 11/28/2022] Open
Abstract
Background: Secreted modular calcium-binding protein 1 (SMOC1) belongs to a family of matricellular proteins; it was involved in embryo development, endothelial cell proliferation, angiogenesis, integrin–matrix interactions, cell adhesion, and regulation of glucose metabolism. Previous studies showed that the expression of SMOC1 was increased in some tumors. However, the prognostic value and the biological function of SMOC1 in tumor remain unclear. Methods: In this study, we explored the expression profile and prognostic value of SMOC1 in pan-cancers, especially glioma, via multiple databases, including Oncomine, Gene Expression Profiling Interactive 2, PrognoScan, Kaplan–Meier plotter, and the Chinese Glioma Genome Atlas database. Furthermore, LinkedOmics was used to identify the genes coexpressed with SMOC1 and to perform Kyoto Encyclopedia of Genes and Genomes pathways and Gene Ontology analysis in low-grade glioma (LGG). Also, the Cancer Single-Cell State Atlas database was used to evaluate the correlation between SMOC1 expression and functional state activities in glioma cells. In addition, the Tumor Immune Estimation Resource and TISIDB databases were used to evaluate the correlations between SMOC1 expression and tumor-infiltrating immune cells in the tumor microenvironment. Results: Compared with normal brain tissues, the expression of SMOC1 was increased in LGG tissues. The higher expression of SMOC1 was significantly correlated with better survival of LGG patients. Additionally, functional analyses showed that the SMOC1 coexpressed genes were inhibited in processes such as response to type I interferon and interferon-gamma, lymphocyte-mediated immunity, leukocyte migration, adaptive immune response, neutrophil-mediated immunity, T cell activation, and pathways including EMC–receptor interaction, Th17 cell differentiation, and leukocyte trans-endothelial migration in LGG. Moreover, the expression of SMOC1 was correlated with stemness, hypoxia, EMT, and metastasis of glioma cells. Additionally, the expression of SMOC1 expression was negatively correlated with levels of infiltrating B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils and dendritic cells, and gene markers of most immune cells in LGG. Conclusion: Our results suggest that SMOC1 could be a potential biomarker to determine prognosis and might play a specific role in the tumor microenvironment of glioma, thereby influencing the development and progression of glioma. These findings provide some new insights for further investigation.
Collapse
Affiliation(s)
- Jing Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu Xia
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingsong Xi
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Walker CJ, Schroeder ME, Aguado BA, Anseth KS, Leinwand LA. Matters of the heart: Cellular sex differences. J Mol Cell Cardiol 2021; 160:42-55. [PMID: 34166708 PMCID: PMC8571046 DOI: 10.1016/j.yjmcc.2021.04.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/12/2021] [Accepted: 04/24/2021] [Indexed: 02/06/2023]
Abstract
Nearly all cardiovascular diseases show sexual dimorphisms in prevalence, presentation, and outcomes. Until recently, most clinical trials were carried out in males, and many animal studies either failed to identify the sex of the animals or combined data obtained from males and females. Cellular sex in the heart is relatively understudied and many studies fail to report the sex of the cells used for in vitro experiments. Moreover, in the small number of studies in which sex is reported, most of those studies use male cells. The observation that cells from males and females are inherently different is becoming increasingly clear - either due to acquired differences from hormones and other factors or due to intrinsic differences in genotype (XX or XY). Because of the likely contribution of cellular sex differences in cardiac health and disease, here, we explore differences in mammalian male and female cells in the heart, including the less-studied non-myocyte cell populations. We discuss how the heart's microenvironment impacts male and female cellular phenotypes and vice versa, including how secretory profiles are dependent on cellular sex, and how hormones contribute to sexually dimorphic phenotypes and cellular functions. Intracellular mechanisms that contribute to sex differences, including gene expression and epigenetic remodeling, are also described. Recent single-cell sequencing studies have revealed unexpected sex differences in the composition of cell types in the heart which we discuss. Finally, future recommendations for considering cellular sex differences in the design of bioengineered in vitro disease models of the heart are provided.
Collapse
Affiliation(s)
- Cierra J Walker
- Materials Science and Engineering Program, University of Colorado, Boulder, CO 80303, United States of America; Interdisciplinary Quantitative Biology, University of Colorado, Boulder, CO 80303, United States of America; BioFrontiers Institute, University of Colorado, Boulder, CO 80303, United States of America
| | - Megan E Schroeder
- Chemical and Biological Engineering Department, University of Colorado, Boulder, CO 80303, United States of America; BioFrontiers Institute, University of Colorado, Boulder, CO 80303, United States of America
| | - Brian A Aguado
- Chemical and Biological Engineering Department, University of Colorado, Boulder, CO 80303, United States of America; BioFrontiers Institute, University of Colorado, Boulder, CO 80303, United States of America
| | - Kristi S Anseth
- Chemical and Biological Engineering Department, University of Colorado, Boulder, CO 80303, United States of America; BioFrontiers Institute, University of Colorado, Boulder, CO 80303, United States of America
| | - Leslie A Leinwand
- BioFrontiers Institute, University of Colorado, Boulder, CO 80303, United States of America; Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, United States of America.
| |
Collapse
|
23
|
Pedrero-Prieto CM, Frontiñán-Rubio J, Alcaín FJ, Durán-Prado M, Peinado JR, Rabanal-Ruiz Y. Biological Significance of the Protein Changes Occurring in the Cerebrospinal Fluid of Alzheimer's Disease Patients: Getting Clues from Proteomic Studies. Diagnostics (Basel) 2021; 11:1655. [PMID: 34573996 PMCID: PMC8467255 DOI: 10.3390/diagnostics11091655] [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: 06/30/2021] [Revised: 08/18/2021] [Accepted: 08/26/2021] [Indexed: 11/16/2022] Open
Abstract
The fact that cerebrospinal fluid (CSF) deeply irrigates the brain together with the relative simplicity of sample extraction from patients make this biological fluid the best target for biomarker discovery in neurodegenerative diseases. During the last decade, biomarker discovery has been especially fruitful for the identification new proteins that appear in the CSF of Alzheimer's disease (AD) patients together with amyloid-β (Aβ42), total tau (T-tau), and phosphorylated tau (P-tau). Thus, several proteins have been already stablished as important biomarkers, due to an increase (i.e., CHI3L1) or a decrease (i.e., VGF) in AD patients' CSF. Notwithstanding this, only a deep analysis of a database generated with all the changes observed in CSF across multiple proteomic studies, and especially those using state-of-the-art methodologies, may expose those components or metabolic pathways disrupted at different levels in AD. Deep comparative analysis of all the up- and down-regulated proteins across these studies revealed that 66% of the most consistent protein changes in CSF correspond to intracellular proteins. Interestingly, processes such as those associated to glucose metabolism or RXR signaling appeared inversely represented in CSF from AD patients in a significant manner. Herein, we discuss whether certain cellular processes constitute accurate indicators of AD progression by examining CSF. Furthermore, we uncover new CSF AD markers, such as ITAM, PTPRZ or CXL16, identified by this study.
Collapse
Affiliation(s)
- Cristina M. Pedrero-Prieto
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, CRIB, University of Castilla-La Mancha (UCLM), Paseo de Moledores SN, 13071 Ciudad Real, Spain; (C.M.P.-P.); (J.F.-R.); (F.J.A.); (M.D.-P.)
- Neuroplasticity and Neurodegeneration Laboratory, Ciudad Real Medical School, CRIB, University of Castilla-La Mancha (UCLM), 13005 Ciudad Real, Spain
| | - Javier Frontiñán-Rubio
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, CRIB, University of Castilla-La Mancha (UCLM), Paseo de Moledores SN, 13071 Ciudad Real, Spain; (C.M.P.-P.); (J.F.-R.); (F.J.A.); (M.D.-P.)
| | - Francisco J. Alcaín
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, CRIB, University of Castilla-La Mancha (UCLM), Paseo de Moledores SN, 13071 Ciudad Real, Spain; (C.M.P.-P.); (J.F.-R.); (F.J.A.); (M.D.-P.)
| | - Mario Durán-Prado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, CRIB, University of Castilla-La Mancha (UCLM), Paseo de Moledores SN, 13071 Ciudad Real, Spain; (C.M.P.-P.); (J.F.-R.); (F.J.A.); (M.D.-P.)
| | - Juan R. Peinado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, CRIB, University of Castilla-La Mancha (UCLM), Paseo de Moledores SN, 13071 Ciudad Real, Spain; (C.M.P.-P.); (J.F.-R.); (F.J.A.); (M.D.-P.)
| | - Yoana Rabanal-Ruiz
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, CRIB, University of Castilla-La Mancha (UCLM), Paseo de Moledores SN, 13071 Ciudad Real, Spain; (C.M.P.-P.); (J.F.-R.); (F.J.A.); (M.D.-P.)
| |
Collapse
|
24
|
Secreted modular calcium-binding protein 1 binds and activates thrombin to account for platelet hyperreactivity in diabetes. Blood 2021; 137:1641-1651. [PMID: 33529332 DOI: 10.1182/blood.2020009405] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
Secreted modular calcium-binding protein 1 (SMOC1) is an osteonectin/SPARC-related matricellular protein, whose expression is regulated by microRNA-223 (miR-223). Given that platelets are rich in miR-223, this study investigated the expression of SMOC1 and its contribution to platelet function. Human and murine platelets expressed SMOC1, whereas platelets from SMOC1+/- mice did not present detectable mature SMOC1 protein. Platelets from SMOC1+/- mice demonstrated attenuated responsiveness to thrombin (platelet neutrophil aggregate formation, aggregation, clot formation, Ca2+ increase, and β3 integrin phosphorylation), whereas responses to other platelet agonists were unaffected. SMOC1 has been implicated in transforming growth factor-β signaling, but no link to this pathway was detected in platelets. Rather, the SMOC1 Kazal domain directly bound thrombin to potentiate its activity in vitro, as well as its actions on isolated platelets. The latter effects were prevented by monoclonal antibodies against SMOC1. Platelets from miR-223-deficient mice expressed high levels of SMOC1 and exhibited hyperreactivity to thrombin that was also reversed by preincubation with monoclonal antibodies against SMOC1. Similarly, SMOC1 levels were markedly upregulated in platelets from individuals with type 2 diabetes, and the SMOC1 antibody abrogated platelet hyperresponsiveness to thrombin. Taken together, we have identified SMOC1 as a novel thrombin-activating protein that makes a significant contribution to the pathophysiological changes in platelet function associated with type 2 diabetes. Thus, strategies that target SMOC1 or its interaction with thrombin may be attractive therapeutic approaches to normalize platelet function in diabetes.
Collapse
|
25
|
Yin GN, Piao S, Liu Z, Wang L, Ock J, Kwon MH, Kim DK, Gho YS, Suh JK, Ryu JK. RNA-sequencing profiling analysis of pericyte-derived extracellular vesicle-mimetic nanovesicles-regulated genes in primary cultured fibroblasts from normal and Peyronie's disease penile tunica albuginea. BMC Urol 2021; 21:103. [PMID: 34362357 PMCID: PMC8344132 DOI: 10.1186/s12894-021-00872-x] [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: 09/04/2020] [Accepted: 07/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Peyronie's disease (PD) is a severe fibrotic disease of the tunica albuginea that causes penis curvature and leads to penile pain, deformity, and erectile dysfunction. The role of pericytes in the pathogenesis of fibrosis has recently been determined. Extracellular vesicle (EV)-mimetic nanovesicles (NVs) have attracted attention regarding intercellular communication between cells in the field of fibrosis. However, the global gene expression of pericyte-derived EV-mimetic NVs (PC-NVs) in regulating fibrosis remains unknown. Here, we used RNA-sequencing technology to investigate the potential target genes regulated by PC-NVs in primary fibroblasts derived from human PD plaque. METHODS Human primary fibroblasts derived from normal and PD patients was cultured and treated with cavernosum pericytes isolated extracellular vesicle (EV)-mimetic nanovesicles (NVs). A global gene expression RNA-sequencing assay was performed on normal fibroblasts, PD fibroblasts, and PD fibroblasts treated with PC-NVs. Reverse transcription polymerase chain reaction (RT-PCR) was used for sequencing data validation. RESULTS A total of 4135 genes showed significantly differential expression in the normal fibroblasts, PD fibroblasts, and PD fibroblasts treated with PC-NVs. However, only 91 contra-regulated genes were detected among the three libraries. Furthermore, 20 contra-regulated genes were selected and 11 showed consistent changes in the RNA-sequencing assay, which were validated by RT-PCR. CONCLUSION The gene expression profiling results suggested that these validated genes may be good targets for understanding potential mechanisms and conducting molecular studies into PD.
Collapse
Affiliation(s)
- Guo Nan Yin
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd St, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea
| | - Shuguang Piao
- Department of Urology at Changhai Hospital Affiliated with the Naval Medicine University, Shanghai, 200433, People's Republic of China
| | - Zhiyong Liu
- Department of Urology at Changhai Hospital Affiliated with the Naval Medicine University, Shanghai, 200433, People's Republic of China
| | - Lei Wang
- Department of Urology at Changhai Hospital Affiliated with the Naval Medicine University, Shanghai, 200433, People's Republic of China
| | - Jiyeon Ock
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd St, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea
| | - Mi-Hye Kwon
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd St, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea
| | - Do-Kyun Kim
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Jeonbuk, 54531, Korea
| | - Yong Song Gho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Kyeongsangbuk-do, 37673, Korea
| | - Jun-Kyu Suh
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd St, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea.
| | - Ji-Kan Ryu
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd St, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea.
| |
Collapse
|
26
|
Finger AM, Jäschke S, Del Olmo M, Hurwitz R, Granada AE, Herzel H, Kramer A. Intercellular coupling between peripheral circadian oscillators by TGF-β signaling. SCIENCE ADVANCES 2021; 7:7/30/eabg5174. [PMID: 34301601 PMCID: PMC8302137 DOI: 10.1126/sciadv.abg5174] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/08/2021] [Indexed: 05/04/2023]
Abstract
Coupling between cell-autonomous circadian oscillators is crucial to prevent desynchronization of cellular networks and disruption of circadian tissue functions. While neuronal oscillators within the mammalian central clock, the suprachiasmatic nucleus, couple intercellularly, coupling among peripheral oscillators is controversial and the molecular mechanisms are unknown. Using two- and three-dimensional mammalian culture models in vitro (mainly human U-2 OS cells) and ex vivo, we show that peripheral oscillators couple via paracrine pathways. We identify transforming growth factor-β (TGF-β) as peripheral coupling factor that mediates paracrine phase adjustment of molecular clocks through transcriptional regulation of core-clock genes. Disruption of TGF-β signaling causes desynchronization of oscillator networks resulting in reduced amplitude and increased sensitivity toward external zeitgebers. Our findings reveal an unknown mechanism for peripheral clock synchrony with implications for rhythmic organ functions and circadian health.
Collapse
Affiliation(s)
- Anna-Marie Finger
- Charité Universitätsmedizin Berlin, Institute for Medical Immunology, Laboratory of Chronobiology, Charitéplatz 1, 10117 Berlin, Germany.
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Sebastian Jäschke
- Charité Universitätsmedizin Berlin, Institute for Medical Immunology, Laboratory of Chronobiology, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Marta Del Olmo
- Charité and Humboldt Universität zu Berlin, Institute for Theoretical Biology, Laboratory of Theoretical Chronobiology, Philippstraße 13, 10115 Berlin, Germany
| | - Robert Hurwitz
- Max Planck Institute for Infection Biology, Biochemistry-Protein Purification Core Facility, Charitéplatz 1, 10117 Berlin, Germany
| | - Adrián E Granada
- Charité-Universitätsmedizin, Charité Comprehensive Cancer Center, Laboratory of Systems Oncology, Charitéplatz 1, 10117 Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center, Partner Site Berlin, 69120, Heidelberg, Germany
| | - Hanspeter Herzel
- Charité and Humboldt Universität zu Berlin, Institute for Theoretical Biology, Laboratory of Theoretical Chronobiology, Philippstraße 13, 10115 Berlin, Germany
| | - Achim Kramer
- Charité Universitätsmedizin Berlin, Institute for Medical Immunology, Laboratory of Chronobiology, Charitéplatz 1, 10117 Berlin, Germany.
- Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
27
|
Wang Y, Gu J, Du A, Zhang S, Deng M, Zhao R, Lu Y, Ji Y, Shao Y, Sun W, Kong X. SPARC-related modular calcium binding 1 regulates aortic valve calcification by disrupting BMPR-II/p-p38 signalling. Cardiovasc Res 2021; 118:913-928. [PMID: 33757126 PMCID: PMC8859632 DOI: 10.1093/cvr/cvab107] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 03/21/2021] [Indexed: 02/05/2023] Open
Abstract
Aims Aortic valve calcification is more prevalent in chronic kidney disease accompanied by hypercalcemia. Secreted protein acidic and rich in cysteine (SPARC)-related modular calcium binding 1 (SMOC1) is a regulator of BMP2 signalling, but the role of SMOC1 in aortic valve calcification under different conditions has not been studied. This study aimed to investigate the roles of SMOC1 in aortic valve calcification under normal and high calcium conditions, focusing on the effects on aortic valve interstitial cells (AVICs). Methods and results SMOC1 was expressed by aortic valve endothelial cells and secreted into the extracellular matrix in non-calcific valves and downregulated in calcific aortic valves. In vitro studies demonstrated that HUVEC secreted SMOC1 could enter the cytoplasm of AVICs. Overexpression of SMOC1 attenuated warfarin-induced AVIC calcification but promoted high calcium/phosphate or vitamin D-induced AVIC and aortic valve calcification by regulating BMP2 signalling both in vitro and in vivo. Co-immunoprecipitation revealed that SMOC1 binds to BMP receptor II (BMPR-II) and inhibits BMP2-induced phosphorylation of p38 (p-p38) via amino acids 372–383 of its EF-hand calcium-binding domain. Inhibition of p-p38 by the p38 inhibitor SB203580 blocked the effects of SMOC1 on BMP2 signalling and AVIC calcification induced by high calcium/phosphate medium. In high-calcium-treated AVICs, SMOC1 lost its ability to bind to BMPR-II, but not to caveolin-1, promoting p-p38 and cell apoptosis due to increased expression of BMPR-II and enhanced endocytosis. Conclusions These observations support that SMOC1 works as a dual-directional modulator of AVIC calcification by regulating p38-dependent BMP2 signalling transduction according to different extracellular calcium concentrations.
Collapse
Affiliation(s)
| | | | | | | | | | - Rong Zhao
- Department of Cardiology, The First People's Hospital of Changzhou, 185 Juqian street, Changzhou, 213004, PR China
| | | | | | - Yongfeng Shao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, PR China
| | | | - Xiangqing Kong
- Department of Cardiology.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 210029, PR China
| |
Collapse
|
28
|
Neutrophil extracellular traps promote fibrous vascular occlusions in chronic thrombosis. Blood 2021; 137:1104-1116. [PMID: 33512471 DOI: 10.1182/blood.2020005861] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
Acute pulmonary embolism generally resolves within 6 months. However, if the thrombus is infected, venous thrombi transform into fibrotic vascular obstructions leading to chronic deep vein thrombosis and/or chronic thromboembolic pulmonary hypertension (CTEPH), but precise mechanisms remain unclear. Neutrophils are crucial in sequestering pathogens; therefore, we investigated the role of neutrophil extracellular traps (NETs) in chronic thrombosis. Because chronic pulmonary thrombotic obstructions are biologically identical to chronic deep venous thrombi, the murine inferior vena cava ligation model was used to study the transformation of acute to chronic thrombus. Mice with staphylococcal infection presented with larger thrombi containing more neutrophils and NETs but less resolution. Targeting NETs with DNase1 diminished fibrosis and promoted thrombus resolution. For translational studies in humans, we focused on patients with CTEPH, a severe type of deep venous and pulmonary artery fibrotic obstruction after thrombosis. Neutrophils, markers of neutrophil activation, and NET formation were increased in CTEPH patients. NETs promoted the differentiation of monocytes to activated fibroblasts with the same cellular phenotype as fibroblasts from CTEPH vascular occlusions. RNA sequencing of fibroblasts isolated from thrombo-endarterectomy specimens and pulmonary artery biopsies revealed transforming growth factor-β (TGF-β) as the central regulator, a phenotype which was replicated in mice with fibroblast-specific TGF-β overactivity. Our findings uncover a role of neutrophil-mediated inflammation to enhance TGF-β signaling, which leads to fibrotic thrombus remodeling. Targeting thrombus NETs with DNases may serve as a new therapeutic concept to treat thrombosis and prevent its sequelae.
Collapse
|
29
|
SMOC1 and IL-4 and IL-13 Cytokines Interfere with Ca 2+ Mobilization in Primary Human Keratinocytes. J Invest Dermatol 2021; 141:1792-1801.e5. [PMID: 33484701 DOI: 10.1016/j.jid.2020.12.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 11/23/2022]
Abstract
Immunoregulatory effects of IL-4 and IL-13 and alterations of keratinocyte (KC) differentiation are important factors in the pathogenesis of atopic dermatitis. This study investigated the role of IL-4 and IL-13 in KC responses to changes in extracellular calcium (Ca2+) and analyzed differentiation signals elicited via a Ca2+ sensor, SMOC1. Real-time dynamics of transmembrane Ca2+ influx were assessed in live KCs by flow cytometry and microscopy. Exposure of KCs to a high Ca2+ environment (1.3 mM) triggered a rapid intracellular Ca2+ influx, whereas IL-4- and IL-13-treated cells exhibited a significant decrease in the peak amplitude of Ca2+ influx (P < 0.01). IL-17A and IL-22 did not elicit such responses. Evaluation of intracellular Ca2+ dynamics by microscopy confirmed these observations and revealed heterogeneity of individual KC responses. IL-4 and IL-13 significantly inhibited the expression of Ca2+-binding protein SMOC1 (P < 0.001). Inhibition of epidermal differentiation markers were also observed in SMOC1 small interfering RNA-transfected KCs. Concurrently, the deletion of SMOC1 increased the amplitude of Ca2+ peak response (P < 0.05). In conclusion, our results provide innovative data that IL-4 and IL-13 regulate KC sensitivity to microenvironmental Ca2+ changes and inhibit Ca2+-induced KC differentiation signals. SMOC1 inhibition by IL-4 and IL-13 alters Ca2+ transport in KCs and inhibits differentiation, suggesting a new target for treatment of atopic dermatitis.
Collapse
|
30
|
DNA Methylation Analysis Identifies Patterns in Progressive Glioma Grades to Predict Patient Survival. Int J Mol Sci 2021; 22:ijms22031020. [PMID: 33498463 PMCID: PMC7864199 DOI: 10.3390/ijms22031020] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/15/2021] [Accepted: 01/17/2021] [Indexed: 12/15/2022] Open
Abstract
DNA methylation is an epigenetic change to the genome that impacts gene activities without modification to the DNA sequence. Alteration in the methylation pattern is a naturally occurring event throughout the human life cycle which may result in the development of diseases such as cancer. In this study, we analyzed methylation data from The Cancer Genome Atlas, under the Lower-Grade Glioma (LGG) and Glioblastoma Multiforme (GBM) projects, to identify methylation markers that exhibit unique changes in DNA methylation pattern along with tumor grade progression, to predict patient survival. We found ten glioma grade-associated Cytosine-phosphate-Guanine (CpG) sites that targeted four genes (SMOC1, KCNA4, SLC25A21, and UPP1) and the methylation pattern is strongly associated with glioma specific molecular alterations, primarily isocitrate dehydrogenase (IDH) mutation and chromosome 1p/19q codeletion. The ten CpG sites collectively distinguished a cohort of diffuse glioma patients with remarkably poor survival probability. Our study highlights genes (KCNA4 and SLC25A21) that were not previously associated with gliomas to have contributed to the poorer patient outcome. These CpG sites can aid glioma tumor progression monitoring and serve as prognostic markers to identify patients diagnosed with less aggressive and malignant gliomas that exhibit similar survival probability to GBM patients.
Collapse
|
31
|
Jensen-Cody SO, Potthoff MJ. Hepatokines and metabolism: Deciphering communication from the liver. Mol Metab 2020; 44:101138. [PMID: 33285302 PMCID: PMC7788242 DOI: 10.1016/j.molmet.2020.101138] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/19/2020] [Accepted: 12/01/2020] [Indexed: 02/09/2023] Open
Abstract
Background The liver is a key regulator of systemic energy homeostasis and can sense and respond to nutrient excess and deficiency through crosstalk with multiple tissues. Regulation of systemic energy homeostasis by the liver is mediated in part through regulation of glucose and lipid metabolism. Dysregulation of either process may result in metabolic dysfunction and contribute to the development of insulin resistance or fatty liver disease. Scope of review The liver has recently been recognized as an endocrine organ that secretes hepatokines, which are liver-derived factors that can signal to and communicate with distant tissues. Dysregulation of liver-centered inter-organ pathways may contribute to improper regulation of energy homeostasis and ultimately metabolic dysfunction. Deciphering the mechanisms that regulate hepatokine expression and communication with distant tissues is essential for understanding inter-organ communication and for the development of therapeutic strategies to treat metabolic dysfunction. Major conclusions In this review, we discuss liver-centric regulation of energy homeostasis through hepatokine secretion. We highlight key hepatokines and their roles in metabolic control, examine the molecular mechanisms of each hepatokine, and discuss their potential as therapeutic targets for metabolic disease. We also discuss important areas of future studies that may contribute to understanding hepatokine signaling under healthy and pathophysiological conditions.
Collapse
Affiliation(s)
- Sharon O Jensen-Cody
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA.
| |
Collapse
|
32
|
Chen S, Zou Q, Chen Y, Kuang X, Wu W, Guo M, Cai Y, Li Q. Regulation of SPARC family proteins in disorders of the central nervous system. Brain Res Bull 2020; 163:178-189. [DOI: 10.1016/j.brainresbull.2020.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
|
33
|
Litviňuková M, Talavera-López C, Maatz H, Reichart D, Worth CL, Lindberg EL, Kanda M, Polanski K, Heinig M, Lee M, Nadelmann ER, Roberts K, Tuck L, Fasouli ES, DeLaughter DM, McDonough B, Wakimoto H, Gorham JM, Samari S, Mahbubani KT, Saeb-Parsy K, Patone G, Boyle JJ, Zhang H, Zhang H, Viveiros A, Oudit GY, Bayraktar OA, Seidman JG, Seidman CE, Noseda M, Hubner N, Teichmann SA. Cells of the adult human heart. Nature 2020; 588:466-472. [PMID: 32971526 PMCID: PMC7681775 DOI: 10.1038/s41586-020-2797-4] [Citation(s) in RCA: 895] [Impact Index Per Article: 179.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Advanced insights into disease mechanisms and therapeutic strategies require a deeper understanding of the molecular processes involved in the healthy heart. Knowledge of the full repertoire of cardiac cells and their gene expression profiles is a fundamental first step in this endeavour. Here, using state-of-the-art analyses of large-scale single-cell and single-nucleus transcriptomes, we characterize six anatomical adult heart regions. Our results highlight the cellular heterogeneity of cardiomyocytes, pericytes and fibroblasts, and reveal distinct atrial and ventricular subsets of cells with diverse developmental origins and specialized properties. We define the complexity of the cardiac vasculature and its changes along the arterio-venous axis. In the immune compartment, we identify cardiac-resident macrophages with inflammatory and protective transcriptional signatures. Furthermore, analyses of cell-to-cell interactions highlight different networks of macrophages, fibroblasts and cardiomyocytes between atria and ventricles that are distinct from those of skeletal muscle. Our human cardiac cell atlas improves our understanding of the human heart and provides a valuable reference for future studies.
Collapse
Affiliation(s)
- Monika Litviňuková
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Carlos Talavera-López
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,EMBL - EBI, Wellcome Genome Campus, Hinxton, UK
| | - Henrike Maatz
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Daniel Reichart
- Department of Genetics, Harvard Medical School, Boston, MA, USA.,Department of Cardiology, University Heart & Vascular Center, University of Hamburg, Hamburg, Germany
| | - Catherine L Worth
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Eric L Lindberg
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Masatoshi Kanda
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Department of Rheumatology and Clinical Immunology, Sapporo Medical University, Sapporo, Japan
| | - Krzysztof Polanski
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Matthias Heinig
- Institute of Computational Biology (ICB), HMGU, Neuherberg, Germany.,Department of Informatics, Technische Universitaet Muenchen (TUM), Munich, Germany
| | - Michael Lee
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Kenny Roberts
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Liz Tuck
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Eirini S Fasouli
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Barbara McDonough
- Department of Genetics, Harvard Medical School, Boston, MA, USA.,Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Joshua M Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Sara Samari
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Krishnaa T Mahbubani
- Department of Surgery, University of Cambridge, NIHR Cambridge Biomedical Centre, Cambridge Biorepository for Translational Medicine, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, NIHR Cambridge Biomedical Centre, Cambridge Biorepository for Translational Medicine, Cambridge, UK
| | - Giannino Patone
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Joseph J Boyle
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Hongbo Zhang
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,Department of Histology and Embryology of Zhongshan School of Medicine, Sun-Yat Sen University, Guangzhou, China
| | - Hao Zhang
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anissa Viveiros
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Omer Ali Bayraktar
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - J G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA. .,Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA. .,Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, London, UK. .,British Heart Foundation Centre of Regenerative Medicine, British Heart Foundation Centre of Research Excellence, Imperial College London, London, UK.
| | - Norbert Hubner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. .,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany. .,Charité-Universitätsmedizin, Berlin, Germany. .,Berlin Institute of Health (BIH), Berlin, Germany.
| | - Sarah A Teichmann
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK. .,Deptartment of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| |
Collapse
|
34
|
Montgomery MK, Bayliss J, Devereux C, Bezawork-Geleta A, Roberts D, Huang C, Schittenhelm RB, Ryan A, Townley SL, Selth LA, Biden TJ, Steinberg GR, Samocha-Bonet D, Meex RCR, Watt MJ. SMOC1 is a glucose-responsive hepatokine and therapeutic target for glycemic control. Sci Transl Med 2020; 12:12/559/eaaz8048. [DOI: 10.1126/scitranslmed.aaz8048] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/03/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022]
Abstract
Intertissue communication is a fundamental feature of metabolic regulation, and the liver is central to this process. We have identified sparc-related modular calcium-binding protein 1 (SMOC1) as a glucose-responsive hepatokine and regulator of glucose homeostasis. Acute intraperitoneal administration of SMOC1 improved glycemic control and insulin sensitivity in mice without changes in insulin secretion. SMOC1 exerted its favorable glycemic effects by inhibiting adenosine 3′,5′-cyclic monophosphate (cAMP)–cAMP-dependent protein kinase (PKA)–cAMP response element–binding protein (CREB) signaling in the liver, leading to decreased gluconeogenic gene expression and suppression of hepatic glucose output. Overexpression of SMOC1 in the liver or once-weekly intraperitoneal injections of a stabilized SMOC1-FC fusion protein induced durable improvements in glucose tolerance and insulin sensitivity indb/dbmice, without adverse effects on adiposity, liver histopathology, or inflammation. Furthermore, circulating SMOC1 correlated with hepatic and systemic insulin sensitivity and was decreased in obese, insulin-resistant humans. Together, these findings identify SMOC1 as a potential pharmacological target for the management of glycemic control in type 2 diabetes.
Collapse
Affiliation(s)
| | - Jacqueline Bayliss
- Department of Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Camille Devereux
- Department of Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - David Roberts
- Department of Physiology, Monash University, Melbourne, VIC 3800, Australia
| | - Cheng Huang
- Proteomics and Metabolomics Facility, Monash University, Melbourne, VIC 3800, Australia
| | - Ralf B. Schittenhelm
- Proteomics and Metabolomics Facility, Monash University, Melbourne, VIC 3800, Australia
| | - Andrew Ryan
- TissuPath, Mount Waverley, VIC 3149, Australia
| | - Scott L. Townley
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men’s Health, Adelaide Medical School, University of Adelaide, SA 5005, Australia
| | - Luke A. Selth
- Dame Roma Mitchell Cancer Research Laboratories and Freemasons Foundation Centre for Men’s Health, Adelaide Medical School, University of Adelaide, SA 5005, Australia
- Flinders Centre for Innovation in Cancer and Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Trevor J. Biden
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Gregory R. Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, the Department of Biochemistry and Biomedical Sciences and the Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Dorit Samocha-Bonet
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW 2052, Australia
| | - Ruth C. R. Meex
- Department of Physiology, Monash University, Melbourne, VIC 3800, Australia
- Department of Human Biology, Maastricht University Medical Centre, Maastricht 6229, Netherlands
| | - Matthew J. Watt
- Department of Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
35
|
Inducers of the endothelial cell barrier identified through chemogenomic screening in genome-edited hPSC-endothelial cells. Proc Natl Acad Sci U S A 2020; 117:19854-19865. [PMID: 32759214 DOI: 10.1073/pnas.1911532117] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The blood-retina barrier and blood-brain barrier (BRB/BBB) are selective and semipermeable and are critical for supporting and protecting central nervous system (CNS)-resident cells. Endothelial cells (ECs) within the BRB/BBB are tightly coupled, express high levels of Claudin-5 (CLDN5), a junctional protein that stabilizes ECs, and are important for proper neuronal function. To identify novel CLDN5 regulators (and ultimately EC stabilizers), we generated a CLDN5-P2A-GFP stable cell line from human pluripotent stem cells (hPSCs), directed their differentiation to ECs (CLDN5-GFP hPSC-ECs), and performed flow cytometry-based chemogenomic library screening to measure GFP expression as a surrogate reporter of barrier integrity. Using this approach, we identified 62 unique compounds that activated CLDN5-GFP. Among them were TGF-β pathway inhibitors, including RepSox. When applied to hPSC-ECs, primary brain ECs, and retinal ECs, RepSox strongly elevated barrier resistance (transendothelial electrical resistance), reduced paracellular permeability (fluorescein isothiocyanate-dextran), and prevented vascular endothelial growth factor A (VEGFA)-induced barrier breakdown in vitro. RepSox also altered vascular patterning in the mouse retina during development when delivered exogenously. To determine the mechanism of action of RepSox, we performed kinome-, transcriptome-, and proteome-profiling and discovered that RepSox inhibited TGF-β, VEGFA, and inflammatory gene networks. In addition, RepSox not only activated vascular-stabilizing and barrier-establishing Notch and Wnt pathways, but also induced expression of important tight junctions and transporters. Taken together, our data suggest that inhibiting multiple pathways by selected individual small molecules, such as RepSox, may be an effective strategy for the development of better BRB/BBB models and novel EC barrier-inducing therapeutics.
Collapse
|
36
|
Zhou M, Haque RU, Dammer EB, Duong DM, Ping L, Johnson ECB, Lah JJ, Levey AI, Seyfried NT. Targeted mass spectrometry to quantify brain-derived cerebrospinal fluid biomarkers in Alzheimer's disease. Clin Proteomics 2020; 17:19. [PMID: 32514259 PMCID: PMC7257173 DOI: 10.1186/s12014-020-09285-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Alzheimer's disease (AD) is the most common cause of dementia, characterized by progressive cognitive decline. Protein biomarkers of AD brain pathology, including β-amyloid and Tau, are reflected in cerebrospinal fluid (CSF), yet the identification of additional biomarkers linked to other brain pathophysiologies remains elusive. We recently reported a multiplex tandem-mass tag (TMT) CSF proteomic analysis of nearly 3000 proteins, following depletion of highly abundant proteins and off-line fractionation, across control and AD cases. Of these, over 500 proteins were significantly increased or decreased in AD, including markers reflecting diverse biological functions in brain. Here, we use a targeted mass spectrometry (MS) approach, termed parallel reaction monitoring (PRM), to quantify select CSF biomarkers without pre-depletion or fractionation to assess the reproducibility of our findings and the specificity of changes for AD versus other causes of cognitive impairment. METHOD We nominated 41 proteins (94 peptides) from the TMT CSF discovery dataset, representing a variety of brain cell-types and biological functions, for label-free PRM analysis in a replication cohort of 88 individuals that included 20 normal controls, 37 clinically diagnosed AD cases and 31 cases with non-AD cognitive impairment. To control for technical variables, isotopically labeled synthetic heavy peptide standards were added into each of the 88 CSF tryptic digests. Furthermore, a peptide pool, representing an equivalent amount of peptide from all samples, was analyzed (n = 10) across each batch. Together, this approach enabled us to assess both the intra- and inter-sample differences in peptide signal response and retention time. RESULTS Despite differences in sample preparation, quantitative MS approaches and patient samples, 25 proteins, including Tau, had a consistent and significant change in AD in both the discovery and replication cohorts. Validated CSF markers with low coefficient of variation included the protein products for neuronal/synaptic (GDA, GAP43, SYN1, BASP1, YWHAB, YWHAZ, UCHL1, STMN1 and MAP1B), glial/inflammation (SMOC1, ITGAM, CHI3L1, SPP1, and CHIT1) and metabolic (PKM, ALDOA and FABP3) related genes. Logistical regression analyses revealed several proteins with high sensitivity and specificity for classifying AD cases from controls and other non-AD dementias. SMOC1, YWHAZ, ALDOA and MAP1B emerged as biomarker candidates that could best discriminate between individuals with AD and non-AD cognitive impairment as well as Tau/β-amyloid ratio. Notably, SMOC1 levels in postmortem brain are highly correlated with AD pathology even in the preclinical stage of disease, indicating that CSF SMOC1 levels reflect underlying brain pathology specific for AD. CONCLUSION Collectively these findings highlight the utility of targeted MS approaches to quantify biomarkers associated with AD that could be used for monitoring disease progression, stratifying patients for clinical trials and measuring therapeutic response.
Collapse
Affiliation(s)
- Maotian Zhou
- Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322 USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Rafi U. Haque
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Eric B. Dammer
- Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322 USA
| | - Duc M. Duong
- Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322 USA
| | - Lingyan Ping
- Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322 USA
| | - Erik C. B. Johnson
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - James J. Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Allan I. Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Nicholas T. Seyfried
- Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322 USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322 USA
| |
Collapse
|
37
|
Secreted modular calcium-binding proteins in pathophysiological processes and embryonic development. Chin Med J (Engl) 2020; 132:2476-2484. [PMID: 31613820 PMCID: PMC6831058 DOI: 10.1097/cm9.0000000000000472] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: Secreted modular calcium-binding proteins (SMOCs) are extracellular glycoproteins of the secreted protein, acidic, and rich in cysteine-related modular calcium-binding protein family and include two isoforms, SMOC1 and SMOC2, in humans. Functionally, SMOCs bind to calcium for various cell functions. In this review, we provided a summary of the most recent advancements in and findings of SMOC1 and SMOC2 in development, homeostasis, and disease states. Data sources: All publications in the PubMed database were searched and retrieved (up to July 24, 2019) using various combinations of keywords searching, including SMOC1, SMOC2, and diseases. Study selection: All original studies and review articles of SMOCs in human diseases and embryo development written in English were retrieved and included. Results: SMOC1 and SMOC2 regulate embryonic development, cell homeostasis, and disease pathophysiology. They play an important role in the regulation of cell cycle progression, cell attachment to the extracellular matrix, tissue fibrosis, calcification, angiogenesis, birth defects, and cancer development. Conclusions: SMOC1 and SMOC2 are critical regulators of many cell biological processes and potential therapeutic targets for the control of human cancers and birth defects.
Collapse
|
38
|
Luo MJ, Rao SS, Tan YJ, Yin H, Hu XK, Zhang Y, Liu YW, Yue T, Chen LJ, Li L, Huang YR, Qian YX, Liu ZZ, Cao J, Wang ZX, Luo ZW, Wang YY, Xia K, Tang SY, Chen CY, Xie H. Fasting before or after wound injury accelerates wound healing through the activation of pro-angiogenic SMOC1 and SCG2. Am J Cancer Res 2020; 10:3779-3792. [PMID: 32206122 PMCID: PMC7069085 DOI: 10.7150/thno.44115] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 02/05/2020] [Indexed: 12/26/2022] Open
Abstract
Healing of the chronic diabetic ulceration and large burns remains a clinical challenge. Therapeutic fasting has been shown to improve health. Our study tested whether fasting facilitates diabetic and burn wound healing and explored the underlying mechanism. Methods: The effects of fasting on diabetic and burn wound healing were evaluated by analyzing the rates of wound closure, re-epithelialization, scar formation, collagen deposition, skin cell proliferation and neovascularization using histological analyses and immunostaining. In vitro functional assays were conducted to assess fasting and refeeding on the angiogenic activities of endothelial cells. Transcriptome sequencing was employed to identify the differentially expressed genes in endothelial cells after fasting treatment and the role of the candidate genes in the fasting-induced promotion of angiogenesis was demonstrated. Results: Two times of 24-h fasting in a week after but especially before wound injury efficiently induced faster wound closure, better epidermal and dermal regeneration, less scar formation and higher level of angiogenesis in mice with diabetic or burn wounds. In vitro, fasting alone by serum deprivation did not increase, but rather reduced the abilities of endothelial cell to proliferate, migrate and form vessel-like tubes. However, subsequent refeeding did not merely rescue, but further augmented the angiogenic activities of endothelial cells. Transcriptome sequencing revealed that fasting itself, but not the following refeeding, induced a prominent upregulation of a variety of pro-angiogenic genes, including SMOC1 (SPARC related modular calcium binding 1) and SCG2 (secretogranin II). Immunofluorescent staining confirmed the increase of SMOC1 and SCG2 expression in both diabetic and burn wounds after fasting treatment. When the expression of SMOC1 or SCG2 was down-regulated, the fasting/refeeding-induced pro-angiogenic effects were markedly attenuated. Conclusion: This study suggests that fasting combined with refeeding, but not fasting solely, enhance endothelial angiogenesis through the activation of SMOC1 and SCG2, thus facilitating neovascularization and rapid wound healing.
Collapse
|
39
|
PRDM16 orchestrates angiogenesis via neural differentiation in the developing brain. Cell Death Differ 2020; 27:2313-2329. [PMID: 32015502 DOI: 10.1038/s41418-020-0504-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis plays crucial roles in maintaining the complex operation of central nervous system (CNS) development. The architecture of communication between neurogenesis and angiogenesis is essential to maintain normal brain development and function. Hence, any disruption of neuron-vascular communications may lead to the pathophysiology of cerebrovascular diseases and blood-brain barrier (BBB) dysfunction. Here we demonstrate that neural differentiation and communication are required for vascular development. Regarding the cellular and molecular mechanism, our results show that PRDM16 activity determines the production of mature neurons and their specific positions in the neocortex. In the cortical plate (CP), aberrant neurons fail to secrete modular calcium-binding protein 1 (SMOC1), an important neuronal signal that participates in neurovascular communication to regulate CNS angiogenesis. Neuronal SMOC1 interacts with TGFBR1 by activating the transcription factors phospho-Smad2/3 to convey intercellular signals to endothelial cells (ECs) in the TGF-β-Smad signaling pathway. Together, our results highlight a crucial coordinated neurovascular development process orchestrated by PRDM16 and reveal the importance of intimate communication for building the neurovascular network during brain development.
Collapse
|
40
|
Bastounis EE, Yeh YT, Theriot JA. Subendothelial stiffness alters endothelial cell traction force generation while exerting a minimal effect on the transcriptome. Sci Rep 2019; 9:18209. [PMID: 31796790 PMCID: PMC6890669 DOI: 10.1038/s41598-019-54336-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/01/2019] [Indexed: 02/07/2023] Open
Abstract
Endothelial cells respond to changes in subendothelial stiffness by altering their migration and mechanics, but whether those responses are due to transcriptional reprogramming remains largely unknown. We measured traction force generation and also performed gene expression profiling for two endothelial cell types grown in monolayers on soft or stiff matrices: primary human umbilical vein endothelial cells (HUVEC) and immortalized human microvascular endothelial cells (HMEC-1). Both cell types respond to changes in subendothelial stiffness by increasing the traction stresses they exert on stiffer as compared to softer matrices, and exhibit a range of altered protein phosphorylation or protein conformational changes previously implicated in mechanotransduction. However, the transcriptome has only a minimal role in this conserved biomechanical response. Only few genes were differentially expressed in each cell type in a stiffness-dependent manner, and none were shared between them. In contrast, thousands of genes were differentially regulated in HUVEC as compared to HMEC-1. HUVEC (but not HMEC-1) upregulate expression of TGF-β2 on stiffer matrices, and also respond to application of exogenous TGF-β2 by enhancing their endogenous TGF-β2 expression and their cell-matrix traction stresses. Altogether, these findings provide insights into the relationship between subendothelial stiffness, endothelial mechanics and variation of the endothelial cell transcriptome, and reveal that subendothelial stiffness, while critically altering endothelial cells’ mechanical behavior, minimally affects their transcriptome.
Collapse
Affiliation(s)
- Effie E Bastounis
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195-1800, USA
| | - Yi-Ting Yeh
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195-1800, USA.
| |
Collapse
|
41
|
Siuda D, Randriamboavonjy V, Fleming I. Regulation of calpain 2 expression by miR-223 and miR-145. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1862:194438. [PMID: 31634637 DOI: 10.1016/j.bbagrm.2019.194438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/26/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022]
Abstract
Calpain 2 (CAPN2) is a Ca2+-dependent cysteine-protease that is involved in different cellular processes. Despite its important role, little is known about how CAPN2 expression is regulated. This study addressed the potential regulation of CAPN2 by microRNAs (miRNAs) in human endothelial cells. Two miRNAs were found to regulate CAPN2 expression by two distinct mechanisms, one direct and the other indirect. MiR-223 directly targeted CAPN2 by binding to the CAPN2 3'-untranslated region. Mir-223 overexpression decreased CAPN2 protein levels in cultured cells and in mice miR-223 antagonism led to an increase in CAPN2 protein in lung tissue. MiR-145 overexpression also decreased CAPN2 expression but did not affect a CAPN2 luciferase construct, indicating that the effect was indirect. MiR-145 targets histone deacetylase (HDAC) 2, and HDAC inhibition transcriptionally regulated CAPN2 expression by hyperacetylation of the promoter of CAPN2 gene and a subsequent decrease in polymerase 2 binding. Indeed, down regulation of HDAC2 by miR-145 not only decreased CAPN2 protein expression and calpain activity, but also protected paxillin against calpain-dependent degradation. Thus, protein levels of CAPN2 are regulated by miR-223, acting directly on the 3'-untranslated region as well as by miR-145, which acts via an increase in HDAC2. ENZYMES: Calpain 2 (EC 3.4.22.53), histone deacetylase 2 (EC 3.5.1.98).
Collapse
Affiliation(s)
- Daniel Siuda
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany; German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, Frankfurt am Main, Germany
| | - Voahanginirina Randriamboavonjy
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany; German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany; German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, Frankfurt am Main, Germany.
| |
Collapse
|
42
|
Role of TGF-Beta1/SMAD2/3 Pathway in Retinal Outer Deep Vascular Plexus and Photoreceptor Damage in Rat 50/10 Oxygen-Induced Retinopathy. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4072319. [PMID: 31240212 PMCID: PMC6556365 DOI: 10.1155/2019/4072319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/28/2019] [Accepted: 04/28/2019] [Indexed: 11/25/2022]
Abstract
In retinopathy of prematurity (ROP), outer deep vascular plexus (oDVP) was the emerging field, and the mechanisms of photoreceptor dysfunction remained to be explored. ODVP and photoreceptors were related, with oDVP being part of the supplier of oxygen and nutrients to photoreceptors, while their possible relationship in ROP was not clear. TGF-beta1 has been reported indispensable in oDVP development and altered in ROP patients and animal models. We hypothesized that the TGF-beta1 alteration in rat 50/10 oxygen-induced retinopathy (OIR) model contributed to oDVP malformation and exerted consequent effects on photoreceptor development. We first explored the profile of oDVP development in rat after birth and compared the expression of TGF-beta1 and pSMAD2/3 in Normoxia and OIR groups. Afterwards, the inhibitor of the pathway, LY364947, was used to establish the OIR, OIR+LY364947, Normoxia, and Normoxia+LY364947 groups. The oDVP and photoreceptor were examined by Isolectin B4 staining, western-blot of CD31 and Rho, and electron microscopy. ODVP sprouted at postnatal day 10 (D10) and reached the edge of retina at D14. The TGF-beta1/SMAD2/3 pathway was compromised during the critical period of oDVP development. The inhibitor simulated the oDVP retardation, pericyte, and photoreceptor malformation in the Normoxia+LY364947 group and might further compromise the development of oDVP and photoreceptor in the OIR+LY364947 group. The inhibition of the TGF-beta1/SMAD2/3 pathway indicated its critical role in oDVP malformation and photoreceptor damage, suggesting a possible therapeutic target of ROP treatment.
Collapse
|
43
|
DeGroot MS, Shi H, Eastman A, McKillop AN, Liu J. The Caenorhabditis elegans SMOC-1 Protein Acts Cell Nonautonomously To Promote Bone Morphogenetic Protein Signaling. Genetics 2019; 211:683-702. [PMID: 30518528 PMCID: PMC6366928 DOI: 10.1534/genetics.118.301805] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 12/04/2018] [Indexed: 01/20/2023] Open
Abstract
Bone morphogenetic protein (BMP) signaling regulates many different developmental and homeostatic processes in metazoans. The BMP pathway is conserved in Caenorhabditis elegans, and is known to regulate body size and mesoderm development. We have identified the C. elegans smoc-1 (Secreted MOdular Calcium-binding protein-1) gene as a new player in the BMP pathway. smoc-1(0) mutants have a small body size, while overexpression of smoc-1 leads to a long body size and increased expression of the RAD-SMAD (reporter acting downstream of SMAD) BMP reporter, suggesting that SMOC-1 acts as a positive modulator of BMP signaling. Using double-mutant analysis, we showed that SMOC-1 antagonizes the function of the glypican LON-2 and acts through the BMP ligand DBL-1 to regulate BMP signaling. Moreover, SMOC-1 appears to specifically regulate BMP signaling without significant involvement in a TGFβ-like pathway that regulates dauer development. We found that smoc-1 is expressed in multiple tissues, including cells of the pharynx, intestine, and posterior hypodermis, and that the expression of smoc-1 in the intestine is positively regulated by BMP signaling. We further established that SMOC-1 functions cell nonautonomously to regulate body size. Human SMOC1 and SMOC2 can each partially rescue the smoc-1(0) mutant phenotype, suggesting that SMOC-1's function in modulating BMP signaling is evolutionarily conserved. Together, our findings highlight a conserved role of SMOC proteins in modulating BMP signaling in metazoans.
Collapse
Affiliation(s)
- Melisa S DeGroot
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853
| | - Herong Shi
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853
| | - Alice Eastman
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853
| | - Alexandra N McKillop
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853
| | - Jun Liu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853
| |
Collapse
|
44
|
The Controversial Role of TGF-β in Neovascular Age-Related Macular Degeneration Pathogenesis. Int J Mol Sci 2018; 19:ijms19113363. [PMID: 30373226 PMCID: PMC6275040 DOI: 10.3390/ijms19113363] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/18/2022] Open
Abstract
The multifunctional transforming growth factors-beta (TGF-βs) have been extensively studied regarding their role in the pathogenesis of neovascular age-related macular degeneration (nAMD), a major cause of severe visual loss in the elderly in developed countries. Despite this, their effect remains somewhat controversial. Indeed, both pro- and antiangiogenic activities have been suggested for TGF-β signaling in the development and progression of nAMD, and opposite therapies have been proposed targeting the inhibition or activation of the TGF-β pathway. The present article summarizes the current literature linking TGF-β and nAMD, and reviews experimental data supporting both pro- and antiangiogenic hypotheses, taking into account the limitations of the experimental approaches.
Collapse
|
45
|
Abstract
SIGNIFICANCE Platelets are anucleate blood cells that are involved in hemostasis and thrombosis. Although no longer able to generate ribonucleic acid (RNA) de novo, platelets contain messenger RNA (mRNA), YRNA fragments, and premature microRNAs (miRNAs) that they inherit from megakaryocytes. Recent Advances: Novel sequencing techniques have helped identify the unexpectedly large number of RNA species present in platelets. Throughout their life time, platelets can process the pre-existing pool of premature miRNA to give the fully functional miRNA that can regulate platelet protein expression and function. CRITICAL ISSUES Platelets make a major contribution to the circulating miRNA pool but platelet activation can have major consequences on Dicer levels and thus miRNA maturation, which has implications for studies that are focused on screening-stored platelets. FUTURE DIRECTIONS It will be important to determine the importance of platelets as donors for miRNA-containing microvesicles that can be taken up and processed by other (particularly vascular) cells, thus contributing to homeostasis as well as disease progression. Antioxid. Redox Signal. 29, 902-921.
Collapse
Affiliation(s)
- Amro Elgheznawy
- 1 Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University , Frankfurt am Main, Germany .,2 German Center for Cardiovascular Research (DZHK) , Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Ingrid Fleming
- 1 Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University , Frankfurt am Main, Germany .,2 German Center for Cardiovascular Research (DZHK) , Partner site Rhein-Main, Frankfurt am Main, Germany
| |
Collapse
|
46
|
Platelet communication with the vascular wall: role of platelet-derived microparticles and non-coding RNAs. Clin Sci (Lond) 2018; 132:1875-1888. [PMID: 30185611 DOI: 10.1042/cs20180580] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 12/24/2022]
Abstract
Platelets play an important role in vascular homeostasis through their interaction with circulating blood cells as well as the vascular wall. Platelet-mediated communication with other cells can take the form of direct cell-cell interactions via membrane receptors or indirectly through the release of different soluble factors stored in their granules as well as through the release of microparticles. The latter carry different proteins and RNAs which are transferred to the target cells. The aim of this review is to discuss the role of platelet-derived factors, adhesion molecules as well as RNAs as mediators of the cross-talk between platelets and the vessel wall.
Collapse
|
47
|
Suppression of SMOC2 reduces bleomycin (BLM)-induced pulmonary fibrosis by inhibition of TGF-β1/SMADs pathway. Biomed Pharmacother 2018; 105:841-847. [DOI: 10.1016/j.biopha.2018.03.058] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 03/10/2018] [Accepted: 03/11/2018] [Indexed: 12/14/2022] Open
|
48
|
Abstract
Neuron-glia antigen 2-expressing glial cells (NG2 glia) serve as oligodendrocyte progenitors during development and adulthood. However, recent studies have shown that these cells represent not only a transitional stage along the oligodendroglial lineage, but also constitute a specific cell type endowed with typical properties and functions. Namely, NG2 glia (or subsets of NG2 glia) establish physical and functional interactions with neurons and other central nervous system (CNS) cell types, that allow them to constantly monitor the surrounding neuropil. In addition to operating as sensors, NG2 glia have features that are expected for active modulators of neuronal activity, including the expression and release of a battery of neuromodulatory and neuroprotective factors. Consistently, cell ablation strategies targeting NG2 glia demonstrate that, beyond their role in myelination, these cells contribute to CNS homeostasis and development. In this review, we summarize and discuss the advancements achieved over recent years toward the understanding of such functions, and propose novel approaches for further investigations aimed at elucidating the multifaceted roles of NG2 glia.
Collapse
|
49
|
Seldin MM, Koplev S, Rajbhandari P, Vergnes L, Rosenberg GM, Meng Y, Pan C, Phuong TMN, Gharakhanian R, Che N, Mäkinen S, Shih DM, Civelek M, Parks BW, Kim ED, Norheim F, Chella Krishnan K, Hasin-Brumshtein Y, Mehrabian M, Laakso M, Drevon CA, Koistinen HA, Tontonoz P, Reue K, Cantor RM, Björkegren JLM, Lusis AJ. A Strategy for Discovery of Endocrine Interactions with Application to Whole-Body Metabolism. Cell Metab 2018; 27:1138-1155.e6. [PMID: 29719227 PMCID: PMC5935137 DOI: 10.1016/j.cmet.2018.03.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 12/14/2017] [Accepted: 03/24/2018] [Indexed: 12/16/2022]
Abstract
Inter-tissue communication via secreted proteins has been established as a vital mechanism for proper physiologic homeostasis. Here, we report a bioinformatics framework using a mouse reference population, the Hybrid Mouse Diversity Panel (HMDP), which integrates global multi-tissue expression data and publicly available resources to identify and functionally annotate novel circuits of tissue-tissue communication. We validate this method by showing that we can identify known as well as novel endocrine factors responsible for communication between tissues. We further show the utility of this approach by identification and mechanistic characterization of two new endocrine factors. Adipose-derived Lipocalin-5 is shown to enhance skeletal muscle mitochondrial function, and liver-secreted Notum promotes browning of white adipose tissue, also known as "beiging." We demonstrate the general applicability of the method by providing in vivo evidence for three additional novel molecules mediating tissue-tissue interactions.
Collapse
Affiliation(s)
- Marcus M Seldin
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Simon Koplev
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Prashant Rajbhandari
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Laurent Vergnes
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Gregory M Rosenberg
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yonghong Meng
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Calvin Pan
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Thuy M N Phuong
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Raffi Gharakhanian
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Nam Che
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Selina Mäkinen
- Department of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Diana M Shih
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mete Civelek
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Brian W Parks
- Department of Nutritional Sciences, University of Wisconsin, Madison, WI, USA
| | - Eric D Kim
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | | | - Margarete Mehrabian
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Heikki A Koistinen
- Department of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Karen Reue
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rita M Cantor
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Johan L M Björkegren
- Department of Genetics and Genomic Sciences, The Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Aldons J Lusis
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA.
| |
Collapse
|
50
|
Yu X, Chen X, Zheng XD, Zhang J, Zhao X, Liu Y, Zhang H, Zhang L, Yu H, Zhang M, Ma C, Hao X, Zhu D. Growth Differentiation Factor 11 Promotes Abnormal Proliferation and Angiogenesis of Pulmonary Artery Endothelial Cells. Hypertension 2018; 71:729-741. [PMID: 29463625 DOI: 10.1161/hypertensionaha.117.10350] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/09/2017] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
Disordered proliferation and angiogenesis of pulmonary artery endothelial cells is an important stage in the development of pulmonary arterial hypertension. Recent studies revealed that GDF11 (growth differentiation factor 11) induces endothelial cells proliferation and migration; however, whether GDF11 is directly involved in the pathogenesis of pulmonary arterial hypertension remains unknown. Here, we found that GDF11 was significantly upregulated and activated in 2 experimental pulmonary arterial hypertension models and cultured pulmonary artery endothelial cells. Genetic ablation of gdf11 in endothelial cells rescued pulmonary arterial hypertension features, as demonstrated by right ventricle hypertrophy, right ventricular systolic pressure, hemodynamics, cardiac function, and vascular remodeling. Moreover, we found that hypoxia significantly increased cell cycle progression, proliferation, migration, adhesion, and tube formation, which were significantly inhibited by GDF11 small interfering RNA. These events could be reproduced using cultured pulmonary artery endothelial cells and were dependent on Smad signaling. Moreover, hypoxia-induced GDF11 expression was regulated by the transcription factor zinc finger protein 740, which assisted RNA polymerase in recognizing and binding to the GDF11 promoter sequence located at a site (-753/-744; CCCCCCCCAC) upstream of the gene. This study identified a novel growth and differentiation factor signaling pathway involved in the zinc finger protein 740/GDF11/transforming growth factor-β receptor I/Smad signaling axis and involved in pulmonary artery endothelial cells proliferation and angiogenesis. These results provide critical insights for the development of novel therapeutic strategies for pulmonary arterial hypertension involving components of the GDF11 signaling system.
Collapse
Affiliation(s)
- Xiufeng Yu
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Xinxin Chen
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Xiao Dong Zheng
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Junting Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Xijuan Zhao
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Ying Liu
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Hongyue Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Lixin Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Hao Yu
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Min Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Cui Ma
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Xuewei Hao
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Daling Zhu
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.).
| |
Collapse
|