1
|
Kang CH, Jung ES, Jung SJ, Han YH, Chae SW, Jeong DY, Kim BC, Lee SO, Yoon SJ. Pasteurized Akkermansia muciniphila HB05 (HB05P) Improves Muscle Strength and Function: A 12-Week, Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients 2024; 16:4037. [PMID: 39683431 PMCID: PMC11643799 DOI: 10.3390/nu16234037] [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: 10/22/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Sarcopenia, a condition marked by muscle wasting due to aging or inactivity, severely affects older populations. We previously showed that pasteurized Akkermansia muciniphila HB05 (HB05P), sourced from the breast milk of healthy Korean women, could mitigate muscle wasting in a dexamethasone-induced rat model. Here, we explored whether the oral administration of HB05P can enhance muscle strength and functionality in elderly individuals. Our objective was to determine if HB05P supplementation could benefit muscle performance in aging adults. METHODS We conducted a 12-week, double-blind, placebo-controlled clinical trial involving 100 individuals aged 60 and above, randomly assigned to receive either HB05P (1.0 × 1010 cells/day) or a placebo. RESULTS The HB05P group showed significant improvements in peak torque and peak torque per body weight of the left leg extensor muscles compared to the placebo group (p = 0.0103 and p = 0.0052). Furthermore, HB05P notably elevated follistatin levels, which counteract myostatin, relative to the placebo group (p = 0.0063). No notable safety concerns arose between the groups. CONCLUSIONS HB05P is a promising postbiotic derived from Akkermansia muciniphila that may enhance muscle strength and be used as a safe postbiotic ingredient of Akkermansia muciniphila to improve muscle health.
Collapse
Affiliation(s)
- Chang-Ho Kang
- HealthBiome, Co., Ltd., 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; (C.-H.K.); (D.Y.J.); (B.-C.K.)
| | - Eun-Soo Jung
- Clinical Trial Center for Functional Foods, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea; (E.-S.J.); (S.-J.J.); (S.-W.C.)
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea;
| | - Su-Jin Jung
- Clinical Trial Center for Functional Foods, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea; (E.-S.J.); (S.-J.J.); (S.-W.C.)
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea;
| | - Yeon-Hee Han
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea;
- Department of Nuclear Medicine, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea
- Department of Nuclear Medicine, Jeonbuk National University Hospital, Jeonju 54896, Republic of Korea
| | - Soo-Wan Chae
- Clinical Trial Center for Functional Foods, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea; (E.-S.J.); (S.-J.J.); (S.-W.C.)
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea;
| | - Do Yeun Jeong
- HealthBiome, Co., Ltd., 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; (C.-H.K.); (D.Y.J.); (B.-C.K.)
| | - Byoung-Chan Kim
- HealthBiome, Co., Ltd., 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; (C.-H.K.); (D.Y.J.); (B.-C.K.)
| | - Seung-Ok Lee
- Clinical Trial Center for Functional Foods, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea; (E.-S.J.); (S.-J.J.); (S.-W.C.)
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea;
- Department of Gastroenterology and Hepatology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea
| | - Sun-Jung Yoon
- Clinical Trial Center for Functional Foods, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea; (E.-S.J.); (S.-J.J.); (S.-W.C.)
- Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea;
- Department of Orthopedic Surgery, Jeonbuk National University Medical School, 567 Baekje-daero, Jeonju 54896, Republic of Korea
| |
Collapse
|
2
|
Sosa J, Oyelakin A, Sinha S. The Reign of Follistatin in Tumors and Their Microenvironment: Implications for Drug Resistance. BIOLOGY 2024; 13:130. [PMID: 38392348 PMCID: PMC10887188 DOI: 10.3390/biology13020130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024]
Abstract
Follistatin (FST) is a potent neutralizer of the transforming growth factor-β superfamily and is associated with normal cellular programs and various hallmarks of cancer, such as proliferation, migration, angiogenesis, and immune evasion. The aberrant expression of FST by solid tumors is a well-documented observation, yet how FST influences tumor progression and therapy response remains unclear. The recent surge in omics data has revealed new insights into the molecular foundation underpinning tumor heterogeneity and its microenvironment, offering novel precision medicine-based opportunities to combat cancer. In this review, we discuss these recent FST-centric studies, thereby offering an updated perspective on the protean role of FST isoforms in shaping the complex cellular ecosystem of tumors and in mediating drug resistance.
Collapse
Affiliation(s)
- Jennifer Sosa
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Akinsola Oyelakin
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA 98101, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA 98101, USA
| | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
3
|
Yang M, Luo S, Yang J, Chen W, He L, Liu D, Zhao L, Wang X. Crosstalk between the liver and kidney in diabetic nephropathy. Eur J Pharmacol 2022; 931:175219. [PMID: 35987257 DOI: 10.1016/j.ejphar.2022.175219] [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: 04/02/2022] [Revised: 08/09/2022] [Accepted: 08/14/2022] [Indexed: 11/26/2022]
Abstract
Diabetic nephropathy (DN) is a serious complication of diabetes, and its pathogenesis has not been fully elucidated. Recently, communication between organs has gradually become a new focus in the study of diseases pathogenesis, and abnormal interorgan communication has been proven to be involved in the occurrence and progression of many diseases. As an important metabolic organ in the human body, the liver plays an important role in maintaining homeostasis in humans. The liver secretes a series of proteins called hepatokines that affect adjacent and distal organs through paracrine or endocrine signaling pathways. In this review, we summarize some of the hepatokines identified to date and describe their roles in DN to discuss the possibility that the liver-renal axis is potentially useful as a therapeutic target for DN. We summarize the important hepatokines identified thus far and discuss their relationship with DN. We propose for the first time that the "liver-renal axis" is a potential therapeutic target in individuals with DN.
Collapse
Affiliation(s)
- Ming Yang
- Department of Nutrition, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shilu Luo
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jinfei Yang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wei Chen
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Liyu He
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Di Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Zhao
- Department of Reproduction and Genetics, The First Affiliated Hospital of Kunming Medical University, China
| | - Xi Wang
- Department of Nutrition, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
4
|
Weng C, Dong H, Mao J, Lang X, Chen J. Characterization and Function of the Interaction of Angiogenin With Alpha-Actinin 2. Front Mol Biosci 2022; 9:837971. [PMID: 35463945 PMCID: PMC9033276 DOI: 10.3389/fmolb.2022.837971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/01/2022] [Indexed: 11/16/2022] Open
Abstract
Angiogenin (ANG) is the first human tumor-derived angiogenic protein, which can promote angiogenesis and tumor growth. In a previous study, we identified alpha-actinin 2 (ACTN2), a cytoskeletal protein, as a direct interacting protein with angiogenin. However, the interaction between ANG and ACTN2 was not characterized in detail, which may provide information on the molecular mechanisms of ANG functions. In this study, we mapped the accurate binding domain and sites in ANG and ACTN2, respectively. In ANG, the residues from 83 to 105 are the smallest motif that can bind to ACTN2. We then use site mutation analysis to identify the precise binding sites of ANG in the interaction and found that the 101st residue arginine (R101) represents the critical residue involved in the ANG–ACTN2 interaction. In ACTN2, the residues from 383 to 632, containing two spectrin domains in the middle of the rod structure of ACTN2, play an important role in the interaction. Furthermore, we validated the interaction of ACTN2-383–632 to ANG by glutathione-S-transferase (GST) pull-down assay. In functional analysis, overexpressed ACTN2-383–632 could impair tumor cell motility observably, including cell migration and invasion. Meanwhile, ACTN2-383–632 overexpression inhibited tumor cell proliferation and survival as well. These data suggest that an excess expression of ACTN2 segment ACTN2-383–632 can inhibit tumor cell motility and proliferation by interfering with the interaction between ANG and ACTN2, which provides a potential mechanism of ANG action in tumor growth and metastasis.
Collapse
Affiliation(s)
- Chunhua Weng
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratoryof Kidney Disease Prevention and Control Technology, Hangzhou, China
- National Key Clinical Department of Kidney Diseases, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- *Correspondence: Chunhua Weng, ; Jianghua Chen,
| | - Haojie Dong
- Department of Hematological Malignancies Translational Science, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, United States
| | - Jiajia Mao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratoryof Kidney Disease Prevention and Control Technology, Hangzhou, China
- National Key Clinical Department of Kidney Diseases, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Xiabing Lang
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratoryof Kidney Disease Prevention and Control Technology, Hangzhou, China
- National Key Clinical Department of Kidney Diseases, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratoryof Kidney Disease Prevention and Control Technology, Hangzhou, China
- National Key Clinical Department of Kidney Diseases, Hangzhou, China
- Institute of Nephrology, Zhejiang University, Hangzhou, China
- Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, China
- *Correspondence: Chunhua Weng, ; Jianghua Chen,
| |
Collapse
|
5
|
Janik S, Bekos C, Hacker P, Raunegger T, Schiefer AI, Müllauer L, Veraar C, Dome B, Klepetko W, Ankersmit HJ, Moser B. Follistatin impacts Tumor Angiogenesis and Outcome in Thymic Epithelial Tumors. Sci Rep 2019; 9:17359. [PMID: 31757999 PMCID: PMC6874542 DOI: 10.1038/s41598-019-53671-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
Tumor angiogenesis is a key factor in the progression of thymic epithelial tumors (TETs). Activin A, a member of the TGFβ family, and its antagonist Follistatin are involved in several human malignancies and angiogenesis. We investigated Activin A and Follistatin in serum and tumor tissue of patients with TETs in relation to microvessel density (MVD), WHO histology classification, tumor stage and outcome. Membranous Activin A expression was detected in all tumor tissues of TETs, while Follistatin staining was found in tumor nuclei and cytoplasm. Patients with TETs presented with significantly higher Activin A and Follistatin serum concentrations compared to healthy volunteers, respectively. Follistatin serum concentrations correlated significantly with tumor stage and decreased to physiologic values after complete tumor resection. Follistatin serum concentrations correlated further with MVD and were associated with significantly worse freedom from recurrence (FFR). Low numbers of immature tumor vessels represented even an independent worse prognostic factor for FFR at multivariable analysis. To conclude, the Activin A - Follistatin axis is involved in the pathogenesis of TETs. Further study of Follistatin and Activin A in TETs is warranted as the molecules may serve as targets to inhibit tumor angiogenesis and tumor progression.
Collapse
Affiliation(s)
- Stefan Janik
- Christian Doppler Laboratory for Diagnosis and Regeneration of Cardiac and Thoracic Diseases, Medical University Vienna, Vienna, Austria.,Department of Otorhinolaryngology, Head and Neck Surgery, Medical University Vienna, Vienna, Austria
| | - Christine Bekos
- Christian Doppler Laboratory for Diagnosis and Regeneration of Cardiac and Thoracic Diseases, Medical University Vienna, Vienna, Austria.,Department of Obstetrics and Gynecology, Division of General Gynecology and Gynecologic Oncology, Medical University Vienna, Vienna, Austria
| | - Philipp Hacker
- Christian Doppler Laboratory for Diagnosis and Regeneration of Cardiac and Thoracic Diseases, Medical University Vienna, Vienna, Austria.,Division of Thoracic Surgery, Department of Surgery, Medical University Vienna, Vienna, Austria
| | - Thomas Raunegger
- Christian Doppler Laboratory for Diagnosis and Regeneration of Cardiac and Thoracic Diseases, Medical University Vienna, Vienna, Austria.,Division of Thoracic Surgery, Department of Surgery, Medical University Vienna, Vienna, Austria
| | - Ana-Iris Schiefer
- Clinical Institute of Pathology, Medical University Vienna, Vienna, Austria
| | - Leonhard Müllauer
- Clinical Institute of Pathology, Medical University Vienna, Vienna, Austria
| | - Cecilia Veraar
- Department of Anaesthesiology, General Intensive Care and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Balazs Dome
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Centre Vienna, Medical University Vienna, Vienna, Austria
| | - Walter Klepetko
- Division of Thoracic Surgery, Department of Surgery, Medical University Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Christian Doppler Laboratory for Diagnosis and Regeneration of Cardiac and Thoracic Diseases, Medical University Vienna, Vienna, Austria.,Division of Thoracic Surgery, Department of Surgery, Medical University Vienna, Vienna, Austria.,Head FFG Project "APOSEC", FOLAB Surgery, Medical University Vienna, Vienna, Austria
| | - Bernhard Moser
- Christian Doppler Laboratory for Diagnosis and Regeneration of Cardiac and Thoracic Diseases, Medical University Vienna, Vienna, Austria. .,Division of Thoracic Surgery, Department of Surgery, Medical University Vienna, Vienna, Austria.
| |
Collapse
|
6
|
Ardisia crispa root hexane fraction suppressed angiogenesis in human umbilical vein endothelial cells (HUVECs) and in vivo zebrafish embryo model. Biomed Pharmacother 2019; 118:109221. [PMID: 31545225 DOI: 10.1016/j.biopha.2019.109221] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 01/08/2023] Open
Abstract
Ardisia crispa Thunb. A. DC. (Primulaceae) has been used extensively as folk-lore medicine in South East Asia including China and Japan to treat various inflammatory related diseases. Ardisia crispa root hexane fraction (ACRH) has been thoroughly studied by our group and it has been shown to exhibit anti-inflammatory, anti-hyperalgesic, anti-arthritic, anti-ulcer, chemoprevention and suppression against inflammation-induced angiogenesis in various animal model. Nevertheless, its effect against human endothelial cells in vitro has not been reported yet. Hence, the aim of the study is to investigate the potential antiangiogenic property of ACRH in human umbilical vein endothelial cells (HUVECs) and zebrafish embryo model. ACRH was separated from the crude ethanolic extract of the plant's root in prior to experimental studies. MTT assay revealed that ACRH exerted a concentration-dependent antiproliferative effect on HUVEC with the IC50 of 2.49 ± 0.04 μg/mL. At higher concentration (10 μg/mL), apoptosis was induced without affecting the cell cycle distribution. Angiogenic properties including migration, invasion and differentiation of HUVECs, evaluated via wound healing, trans-well invasion and tube formation assay respectively, were significantly suppressed by ACRH in a concentration-dependent manner. Noteworthily, significant antiangiogenic effects were observed even at the lowest concentration used (0.1 μg/mL). Expression of proMMP-2, vascular endothelial growth factor (VEGF)-C, VEGF-D, Angiopoietin-2, fibroblast growth factor (FGF)-1, FGF-2, Follistatin, and hepatocyte growth factor (HGF) were significantly reduced in various degrees by ACRH. The ISV formation in zebrafish embryo was significantly suppressed by ACRH at the concentration of 5 μg/mL. These findings revealed the potential of ACRH as antiangiogenic agent by suppressing multiple proangiogenic proteins. Thus, it can be further verified via the transcription of these proteins from their respective DNA, in elucidating their exact pathways.
Collapse
|
7
|
Liang X, Xie R, Su J, Ye B, Wei S, Liang Z, Bai R, Chen Z, Li Z, Gao X. Inhibition of RNA polymerase III transcription by Triptolide attenuates colorectal tumorigenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:217. [PMID: 31122284 PMCID: PMC6533717 DOI: 10.1186/s13046-019-1232-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/15/2019] [Indexed: 12/31/2022]
Abstract
Background Upregulation of RNA polymerase (Pol) III products, including tRNAs and 5S rRNA, in tumor cells leads to enhanced protein synthesis and tumor formation, making it a potential target for cancer treatment. In this study, we evaluated the inhibition of Pol III transcription by triptolide and the anti-cancer effect of this drug in colorectal tumorigenesis. Methods The effect of triptolide on colorectal cancer development was assessed in colorectal cancer mouse models, 3D organoids, and cultured cells. Colorectal cancer cells were treated with triptolide. Pol III transcription was measured by real-time quantitative polymerase chain reaction (PCR). The formation of TFIIIB, a multi-subunit transcription factor for Pol III, was determined by chromatin immunoprecipitation (ChIP), co-immunoprecipitation (Co-IP), and fluorescence resonance energy transfer (FRET). Results Triptolide reduced both tumor number and tumor size in adenomatous polyposis coli (Apc) mutated (ApcMin/+) mice as well as AOM/DSS-induced mice. Moreover, triptolide effectively inhibited colorectal cancer cell proliferation, colony formation, and organoid growth in vitro, which was associated with decreased Pol III target genes. Mechanistically, triptolide treatment blocked TBP/Brf1interaction, leading to the reduced formation of TFIIIB at the promoters of tRNAs and 5S rRNA. Conclusions Together, our data suggest that inhibition of Pol III transcription with existing drugs such as triptolide provides a new avenue for developing novel therapies for colorectal cancer. Electronic supplementary material The online version of this article (10.1186/s13046-019-1232-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xia Liang
- Medical Research Institute, & Guangdong Women and Children's Disease Precision Diagnosis and Treatment Engineering Technology Research Center, Baoan Maternal and Child Health Hospital, Jinan University, Shenzhen, 518102, China
| | - Renxiang Xie
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jinfeng Su
- Medical Research Institute, & Guangdong Women and Children's Disease Precision Diagnosis and Treatment Engineering Technology Research Center, Baoan Maternal and Child Health Hospital, Jinan University, Shenzhen, 518102, China
| | - Bingqi Ye
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Saisai Wei
- Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhibing Liang
- Medical Research Institute, & Guangdong Women and Children's Disease Precision Diagnosis and Treatment Engineering Technology Research Center, Baoan Maternal and Child Health Hospital, Jinan University, Shenzhen, 518102, China
| | - Rongpan Bai
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhanghui Chen
- Affiliated Central People's Hospital of Zhanjiang, Guangdong Medical University, Zhanjiang, 524045, China
| | - Zhongxiang Li
- Medical Research Institute, & Guangdong Women and Children's Disease Precision Diagnosis and Treatment Engineering Technology Research Center, Baoan Maternal and Child Health Hospital, Jinan University, Shenzhen, 518102, China.
| | - Xiangwei Gao
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
8
|
Fahmy-Garcia S, Farrell E, Witte-Bouma J, Robbesom-van den Berge I, Suarez M, Mumcuoglu D, Walles H, Kluijtmans SGJM, van der Eerden BCJ, van Osch GJVM, van Leeuwen JPTM, van Driel M. Follistatin Effects in Migration, Vascularization, and Osteogenesis in vitro and Bone Repair in vivo. Front Bioeng Biotechnol 2019; 7:38. [PMID: 30881954 PMCID: PMC6405513 DOI: 10.3389/fbioe.2019.00038] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/13/2019] [Indexed: 12/16/2022] Open
Abstract
The use of biomaterials and signaling molecules to induce bone formation is a promising approach in the field of bone tissue engineering. Follistatin (FST) is a glycoprotein able to bind irreversibly to activin A, a protein that has been reported to inhibit bone formation. We investigated the effect of FST in critical processes for bone repair, such as cell recruitment, osteogenesis and vascularization, and ultimately its use for bone tissue engineering. In vitro, FST promoted mesenchymal stem cell (MSC) and endothelial cell (EC) migration as well as essential steps in the formation and expansion of the vasculature such as EC tube-formation and sprouting. FST did not enhance osteogenic differentiation of MSCs, but increased committed osteoblast mineralization. In vivo, FST was loaded in an in situ gelling formulation made by alginate and recombinant collagen-based peptide microspheres and implanted in a rat calvarial defect model. Two FST variants (FST288 and FST315) with major differences in their affinity to cell-surface proteoglycans, which may influence their effect upon in vivo bone repair, were tested. In vitro, most of the loaded FST315 was released over 4 weeks, contrary to FST288, which was mostly retained in the biomaterial. However, none of the FST variants improved in vivo bone healing compared to control. These results demonstrate that FST enhances crucial processes needed for bone repair. Further studies need to investigate the optimal FST carrier for bone regeneration.
Collapse
Affiliation(s)
- Shorouk Fahmy-Garcia
- Department of Orthopedics, Erasmus MC, University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | | | - Melva Suarez
- Institute of Tissue Engineering and Regenerative Medicine, Julius-Maximillians University Würzburg, Würzburg, Germany
| | - Didem Mumcuoglu
- Department of Orthopedics, Erasmus MC, University Medical Center, Rotterdam, Netherlands.,Fujifilm Manufacturing Europe B.V., Tilburg, Netherlands
| | - Heike Walles
- Institute of Tissue Engineering and Regenerative Medicine, Julius-Maximillians University Würzburg, Würzburg, Germany
| | | | - Bram C J van der Eerden
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Gerjo J V M van Osch
- Department of Orthopedics, Erasmus MC, University Medical Center, Rotterdam, Netherlands.,Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | | | - Marjolein van Driel
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
9
|
Hoda MA, Rozsas A, Lang E, Klikovits T, Lohinai Z, Torok S, Berta J, Bendek M, Berger W, Hegedus B, Klepetko W, Renyi-Vamos F, Grusch M, Dome B, Laszlo V. High circulating activin A level is associated with tumor progression and predicts poor prognosis in lung adenocarcinoma. Oncotarget 2017; 7:13388-99. [PMID: 26950277 PMCID: PMC4924649 DOI: 10.18632/oncotarget.7796] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/09/2016] [Indexed: 12/22/2022] Open
Abstract
Activin A (ActA)/follistatin (FST) signaling has been shown to be deregulated in different tumor types including lung adenocarcinoma (LADC). Here, we report that serum ActA protein levels are significantly elevated in LADC patients (n=64) as compared to controls (n=46, p=0.015). ActA levels also correlated with more advanced disease stage (p<0.0001) and T (p=0.0035) and N (p=0.0002) factors. M1 patients had significantly higher ActA levels than M0 patients (p<0.001). High serum ActA level was associated with poor overall survival (p<0.0001) and was confirmed as an independent prognostic factor (p=0.004). Serum FST levels were increased only in female LADC patients (vs. female controls, p=0.031). Two out of five LADC cell lines secreted biologically active ActA, while FST was produced in all of them. Transcripts of both type I and II ActA receptors were detected in all five LADC cell lines. In conclusion, our study does not only suggest that measuring blood ActA levels in LADC patients might improve the prediction of prognosis, but also indicates that this parameter might be a novel non-invasive biomarker for identifying LADC patients with organ metastases.
Collapse
Affiliation(s)
- Mir Alireza Hoda
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria.,Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Anita Rozsas
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria.,National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Elisabeth Lang
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Thomas Klikovits
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Zoltan Lohinai
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Szilvia Torok
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Judit Berta
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Matyas Bendek
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Walter Berger
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Balazs Hegedus
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria.,MTA-SE Molecular Oncology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Walter Klepetko
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Ferenc Renyi-Vamos
- Department of Thoracic Surgery, National Institute of Oncology and Semmelweis University, Budapest, Hungary
| | - Michael Grusch
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Balazs Dome
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria.,National Koranyi Institute of Pulmonology, Budapest, Hungary.,Department of Thoracic Surgery, National Institute of Oncology and Semmelweis University, Budapest, Hungary.,Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Viktoria Laszlo
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
10
|
Li S, Chen Y, Sun D, Bai R, Gao X, Yang Y, Sheng J, Xu Z. Angiogenin Prevents Progranulin A9D Mutation-Induced Neuronal-Like Cell Apoptosis Through Cleaving tRNAs into tiRNAs. Mol Neurobiol 2017; 55:1338-1351. [DOI: 10.1007/s12035-017-0396-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 01/10/2017] [Indexed: 12/20/2022]
|
11
|
Shi L, Resaul J, Owen S, Ye L, Jiang WG. Clinical and Therapeutic Implications of Follistatin in Solid Tumours. Cancer Genomics Proteomics 2017; 13:425-435. [PMID: 27807065 DOI: 10.21873/cgp.20005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 10/05/2016] [Indexed: 12/20/2022] Open
Abstract
Follistatin (FST), as a single-chain glycosylated protein, has two major isoforms, FST288 and FST315. The FST315 isoform is the predominant form whilst the FST288 variant accounts for less than 5% of the encoded mRNA. FST is differentially expressed in human tissues and aberrant expression has been observed in a variety of solid tumours, including gonadal, gastric and lung cancer, hepatocellular carcinoma, basal cell carcinoma and melanoma. Based on the current evidence, FST is an antagonist of transforming growth factor beta family members, such as activin and bone morphogenetic proteins (BMPs). FST plays a role in tumourigenesis, metastasis and angiogenesis of solid tumours through its interaction with activin and BMPs, thus resulting in pathophysiological function. In terms of diagnosis, prognosis and therapy, FST has shown strong promise. Through a better understanding of its biological functions, potential clinical applications may yet emerge.
Collapse
Affiliation(s)
- Lei Shi
- Urology Department, Yantai Yu Huang Ding Hospital, Yantai, Shandong Province, P.R. China.,Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K
| | - Jeyna Resaul
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K
| | - Sioned Owen
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K.
| |
Collapse
|
12
|
Magrì A, Munzone A, Peana M, Medici S, Zoroddu MA, Hansson O, Satriano C, Rizzarelli E, La Mendola D. Coordination Environment of Cu(II) Ions Bound to N-Terminal Peptide Fragments of Angiogenin Protein. Int J Mol Sci 2016; 17:ijms17081240. [PMID: 27490533 PMCID: PMC5000638 DOI: 10.3390/ijms17081240] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/21/2016] [Accepted: 07/22/2016] [Indexed: 12/11/2022] Open
Abstract
Angiogenin (Ang) is a potent angiogenic factor, strongly overexpressed in patients affected by different types of cancers. The specific Ang cellular receptors have not been identified, but it is known that Ang-actin interaction induces changes both in the cell cytoskeleton and in the extracellular matrix. Most in vitro studies use the recombinant form (r-Ang) instead of the form that is normally present in vivo ("wild-type", wt-Ang). The first residue of r-Ang is a methionine, with a free amino group, whereas wt-Ang has a glutamic acid, whose amino group spontaneously cyclizes in the pyro-glutamate form. The Ang biological activity is influenced by copper ions. To elucidate the role of such a free amino group on the protein-copper binding, we scrutinized the copper(II) complexes with the peptide fragments Ang(1-17) and AcAng(1-17), which encompass the sequence 1-17 of angiogenin (QDNSRYTHFLTQHYDAK-NH₂), with free amino and acetylated N-terminus, respectively. Potentiometric, ultraviolet-visible (UV-vis), nuclear magnetic resonance (NMR) and circular dichroism (CD) studies demonstrate that the two peptides show a different metal coordination environment. Confocal microscopy imaging of neuroblastoma cells with the actin staining supports the spectroscopic results, with the finding of different responses in the cytoskeleton organization upon the interaction, in the presence or not of copper ions, with the free amino and the acetylated N-terminus peptides.
Collapse
Affiliation(s)
- Antonio Magrì
- Institute of Biostructures and Bioimages, National Council of Research ( CNR), Via P. Gaifami 18, 95126 Catania, Italy.
| | - Alessia Munzone
- Department of Chemical Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.
| | - Massimiliano Peana
- Department of Chemistry and Pharmacy, University of Sassari, Via Vienna 2, 07100 Sassari, Italy.
| | - Serenella Medici
- Department of Chemistry and Pharmacy, University of Sassari, Via Vienna 2, 07100 Sassari, Italy.
| | - Maria Antonietta Zoroddu
- Department of Chemistry and Pharmacy, University of Sassari, Via Vienna 2, 07100 Sassari, Italy.
| | - Orjan Hansson
- Department of Chemistry and Molecular Biology, University of Gothenburg, Medicinaregatan 9C, 41390 Göteborg, Sweden.
| | - Cristina Satriano
- Department of Chemical Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.
| | - Enrico Rizzarelli
- Institute of Biostructures and Bioimages, National Council of Research ( CNR), Via P. Gaifami 18, 95126 Catania, Italy.
- Department of Chemical Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy.
| |
Collapse
|
13
|
Sheng J, Xu Z. Three decades of research on angiogenin: a review and perspective. Acta Biochim Biophys Sin (Shanghai) 2016; 48:399-410. [PMID: 26705141 DOI: 10.1093/abbs/gmv131] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/23/2015] [Indexed: 01/17/2023] Open
Abstract
As a member of the vertebrate-specific secreted ribonucleases, angiogenin (ANG) was first isolated and identified solely by its ability to induce new blood vessel formation, and now, it has been recognized to play important roles in various physiological and pathological processes through regulating cell proliferation, survival, migration, invasion, and/or differentiation. ANG exhibits very weak ribonucleolytic activity that is critical for its biological functions, and exerts its functions through activating different signaling transduction pathways in different target cells. A series of recent studies have indicated that ANG contributes to cellular nucleic acid metabolism. Here, we comprehensively review the results of studies regarding the structure, mechanism, and function of ANG over the past three decades. Moreover, current problems and future research directions of ANG are discussed. The understanding of the function and mechanism of ANG in a wide context will help to better delineate its roles in diseases, especially in cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jinghao Sheng
- Institute of Environmental Health, Zhejiang University School of Public Health, Hangzhou 310058, China Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhengping Xu
- Institute of Environmental Health, Zhejiang University School of Public Health, Hangzhou 310058, China Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou 310003, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
14
|
Wekesa A, Harrison M, Watson RW. Physical activity and its mechanistic effects on prostate cancer. Prostate Cancer Prostatic Dis 2015; 18:197-207. [PMID: 25800589 DOI: 10.1038/pcan.2015.9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 02/04/2015] [Accepted: 02/06/2015] [Indexed: 12/22/2022]
Abstract
Beneficial effects of physical activity have been illustrated in numerous aspects of health. With the increasing incidence of prostate cancer and changes in physical activity of men, understanding the link between the two has important implications for changing this cancer burden. Both positive and negative associations between physical activity and prostate cancer have been previously demonstrated in observational epidemiological studies. Elucidating the biological mechanisms would lead to a better understanding of how physical activity influences the progression of prostate cancer. This review was undertaken to: (1) identify evidence in literature that demonstrates the effects of physical activity on skeletal muscle secretomes, (2) indicate the plausible signaling pathways these proteins might activate, and (3) identify evidence in literature that demonstrates the roles of the signaling pathways in prostate cancer progression and regression. We also discuss proposed biological mechanisms and signaling pathways by which physical activity may prevent the development and progression of prostate cancer. We discuss proteins involved in the normal and aberrant growth and development of the prostate gland that may be affected by physical activity. We further identify future directions for research, including a better understanding of the biological mechanisms, the need to standardize physical activity and identify mechanistic end points of physical activity that can then be correlated with outcomes.
Collapse
Affiliation(s)
- A Wekesa
- UCD School of Medicine and Medical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - M Harrison
- Department of Health, Sport and Exercise Science, Waterford Institute of Technology, Waterford, Ireland
| | - R W Watson
- UCD School of Medicine and Medical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
15
|
Aguirre Palma LM, Gehrke I, Kreuzer KA. Angiogenic factors in chronic lymphocytic leukaemia (CLL): Where do we stand? Crit Rev Oncol Hematol 2014; 93:225-36. [PMID: 25459668 DOI: 10.1016/j.critrevonc.2014.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 07/23/2014] [Accepted: 10/01/2014] [Indexed: 01/09/2023] Open
Abstract
The role of angiogenesis in haematological malignancies such as chronic lymphocytic leukaemia (CLL) is difficult to envision, because leukaemia cells are not dependent on a network of blood vessels to support basic physiological requirements. Regardless, CLL cells secrete high levels of major angiogenic factors, such as vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and platelet derived growth factor (PDGF). Nonetheless, it remains unclear how most angiogenic factors regulate accumulation and delayed apoptosis of CLL cells. Angiogenic factors such as leptin, granulocyte colony-stimulating factor (G-CSF), follistatin, angiopoietin-1 (Ang1), angiogenin (ANG), midkine (MK), pleiotrophin (PTN), progranulin (PGRN), proliferin (PLF), placental growth factor (PIGF), and endothelial locus-1 (Del-1), represent novel therapeutic targets of future CLL research but have remained widely overlooked. This review aims to outline our current understanding of angiogenic growth factors and their relationship with CLL, a still uncured haematopoietic malignancy.
Collapse
Affiliation(s)
| | - Iris Gehrke
- Manitoba Institute of Cell Biology, University of Manitoba, Winnipeg, MB, Canada.
| | - Karl-Anton Kreuzer
- Department I of Internal Medicine, University of Cologne, Cologne, Germany.
| |
Collapse
|
16
|
Gao X, Dong H, Lin C, Sheng J, Zhang F, Su J, Xu Z. Reduction of AUF1-mediated follistatin mRNA decay during glucose starvation protects cells from apoptosis. Nucleic Acids Res 2014; 42:10720-30. [PMID: 25159612 PMCID: PMC4176339 DOI: 10.1093/nar/gku778] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Follistatin (FST) performs several vital functions in the cells, including protection from apoptosis during stress. The expression of FST is up-regulated in response to glucose deprivation by an unknown mechanism. We herein showed that the induction of FST by glucose deprivation was due to an increase in the half-life of its mRNA. We further identified an AU-rich element (ARE) in the 3′UTR of FST mRNA that mediated its decay. The expression of FST was elevated after knocking down AUF1 and reduced when AUF1 was further expressed. In vitro binding assays and RNA pull-down assays revealed that AUF1 interacted with FST mRNA directly via its ARE. During glucose deprivation, a majority of AUF1 shuttled from cytoplasm to nucleus, resulting in dissociation of AUF1 from FST mRNA and thus stabilization of FST mRNA. Finally, knockdown of AUF1 decreased whereas overexpression of AUF1 increased glucose deprivation-induced apoptosis. The apoptosis promoting effect of AUF1 was eliminated in FST expressing cells. Collectively, this study provided evidence that AUF1 is a negative regulator of FST expression and participates in the regulation of cell survival under glucose deprivation.
Collapse
Affiliation(s)
- Xiangwei Gao
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Haojie Dong
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Chen Lin
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Jinghao Sheng
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Fan Zhang
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Jinfeng Su
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Zhengping Xu
- Institute of Environmental Medicine, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China Program in Molecular Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou 310058, China
| |
Collapse
|
17
|
Tomoda T, Nouso K, Miyahara K, Kobayashi S, Kinugasa H, Toyosawa J, Hagihara H, Kuwaki K, Onishi H, Nakamura S, Ikeda F, Miyake Y, Shiraha H, Takaki A, Yamamoto K. Prognotic impact of serum follistatin in patients with hepatocellular carcinoma. J Gastroenterol Hepatol 2013; 28:1391-1396. [PMID: 23432377 DOI: 10.1111/jgh.12167] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/04/2013] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIM Follistatin (FST) is a glycoprotein expressed in most organs, which interacts with activins or other members of the transforming growth factor beta family. Recently, several reports have shown that FST regulates a variety of processes during tumor progression. Here, serum FST in patients with liver diseases was measured, and its clinical utility as a biomarker was assessed. METHODS Serum was collected from 162 patients (91 hepatocellular carcinoma [HCC], 43 liver cirrhosis, and 28 chronic hepatitis) as well as from 16 healthy volunteers. FST was quantified by enzyme-linked immunosorbent assays, and levels were compared with clinical parameters including survival of the HCC patients. RESULTS Median serum FST levels in HCC, liver cirrhosis, chronic hepatitis, and healthy volunteers were 1168, 1606, 1324, and 1661 pg/mL, respectively, not significantly different. In HCC patients, higher serum FST was associated with greater age, hepatitis C virus antibody-negativity, large tumor size, g-glutamyl transpeptidase, des-gamma carboxyprothrombin and presence of portal vein tumor thrombus. Survival of HCC patients with high FST levels was significantly shorter than for those with low levels (P = 0.004). Multivariate analysis revealed that in addition to large tumor size and presence of portal vein thrombus, high FST levels were independently correlated with poor prognosis (hazard ratio = 2.41, 95% confidence interval = 1.16-5.00, P = 0.02). CONCLUSIONS Serum FST levels are significantly associated with HCC prognosis and could represent a predictive biomarker in this disease.
Collapse
Affiliation(s)
- Takeshi Tomoda
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama City, Okayama, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Raven LA, Cocks BG, Pryce JE, Cottrell JJ, Hayes BJ. Genes of the RNASE5 pathway contain SNP associated with milk production traits in dairy cattle. Genet Sel Evol 2013; 45:25. [PMID: 23865486 PMCID: PMC3733968 DOI: 10.1186/1297-9686-45-25] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 06/26/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Identification of the processes and mutations responsible for the large genetic variation in milk production among dairy cattle has proved challenging. One approach is to identify a biological process potentially involved in milk production and to determine the genetic influence of all the genes included in the process or pathway. Angiogenin encoded by angiogenin, ribonuclease, RNase A family 5 (RNASE5) is relatively abundant in milk, and has been shown to regulate protein synthesis and act as a growth factor in epithelial cells in vitro. However, little is known about the role of angiogenin in the mammary gland or if the polymorphisms present in the bovine RNASE5 gene are associated with lactation and milk production traits in dairy cattle. Given the high economic value of increased protein in milk, we have tested the hypothesis that RNASE5 or genes in the RNASE5 pathway are associated with milk production traits. First, we constructed a "RNASE5 pathway" based on upstream and downstream interacting genes reported in the literature. We then tested SNP in close proximity to the genes of this pathway for association with milk production traits in a large dairy cattle dataset. RESULTS The constructed RNASE5 pathway consisted of 11 genes. Association analysis between SNP in 1 Mb regions surrounding these genes and milk production traits revealed that more SNP than expected by chance were associated with milk protein percent (P < 0.05 significance). There was no significant association with other traits such as milk fat content or fertility. CONCLUSIONS These results support a role for the RNASE5 pathway in milk production, specifically milk protein percent, and indicate that polymorphisms in or near these genes explain a proportion of the variation for this trait. This method provides a novel way of understanding the underlying biology of lactation with implications for milk production and can be applied to any pathway or gene set to test whether they are responsible for the variation of complex traits.
Collapse
Affiliation(s)
- Lesley-Ann Raven
- Biosciences Research Division, Department of Primary Industries Victoria, 5 Ring Rd, Bundoora 3086, Australia.
| | | | | | | | | |
Collapse
|
19
|
Campanati A, Moroncini G, Ganzetti G, Pozniak K, Goteri G, Giuliano A, Martina E, Liberati G, Ricotti F, Gabrielli A, Offidani A. Adalimumab Modulates Angiogenesis in Psoriatic Skin. EUR J INFLAMM 2013. [DOI: 10.1177/1721727x1301100218] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- A. Campanati
- Dermatological Clinic, Department of Clinical and Molecular Medicine, Ancona Hospital, Polytechnic University of Marche Region, Ancona, Italy
| | - G. Moroncini
- Internal Medicine, Department of Clinical and Molecular Medicine, Ancona Hospital, Polytechnic University of Marche Region, Ancona, Italy
| | - G. Ganzetti
- Dermatological Clinic, Department of Clinical and Molecular Medicine, Ancona Hospital, Polytechnic University of Marche Region, Ancona, Italy
| | - K.N. Pozniak
- Internal Medicine, Department of Clinical and Molecular Medicine, Ancona Hospital, Polytechnic University of Marche Region, Ancona, Italy
| | - G. Goteri
- Anatomic Pathology Institute, Ancona Hospital, Department of Neurosciences, Polytechnic University of Marche Region, Ancona, Italy
| | - A. Giuliano
- Dermatological Clinic, Department of Clinical and Molecular Medicine, Ancona Hospital, Polytechnic University of Marche Region, Ancona, Italy
| | - E. Martina
- Dermatological Clinic, Department of Clinical and Molecular Medicine, Ancona Hospital, Polytechnic University of Marche Region, Ancona, Italy
| | - G. Liberati
- Dermatological Clinic, Department of Clinical and Molecular Medicine, Ancona Hospital, Polytechnic University of Marche Region, Ancona, Italy
| | - F. Ricotti
- Dermatological Clinic, Department of Clinical and Molecular Medicine, Ancona Hospital, Polytechnic University of Marche Region, Ancona, Italy
| | - A. Gabrielli
- Internal Medicine, Department of Clinical and Molecular Medicine, Ancona Hospital, Polytechnic University of Marche Region, Ancona, Italy
| | - A. Offidani
- Dermatological Clinic, Department of Clinical and Molecular Medicine, Ancona Hospital, Polytechnic University of Marche Region, Ancona, Italy
| |
Collapse
|
20
|
Sepporta MV, Tumminello FM, Flandina C, Crescimanno M, Giammanco M, La Guardia M, di Majo D, Leto G. Follistatin as potential therapeutic target in prostate cancer. Target Oncol 2013; 8:215-23. [PMID: 23456439 DOI: 10.1007/s11523-013-0268-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 02/05/2013] [Indexed: 01/04/2023]
Abstract
Follistatin is a single-chain glycosylated protein whose primary function consists in binding and neutralizing some members of the transforming growth factor-β superfamily such as activin and bone morphogenic proteins. Emerging evidence indicates that this molecule may also play a role in the malignant progression of several human tumors including prostate cancer. In particular, recent findings suggest that, in this tumor, follistatin may also contribute to the formation of bone metastasis through multiple mechanisms, some of which are not related to its specific activin or bone morphogenic proteins' inhibitory activity. This review provides insight into the most recent advances in understanding the role of follistatin in the prostate cancer progression and discusses the clinical and therapeutic implications related to these findings.
Collapse
Affiliation(s)
- Maria Vittoria Sepporta
- Operative Unit of Physiology and Pharmacology, University of Palermo, via Augusto Elia, 3, 90127, Palermo, Italy
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Karve TM, Preet A, Sneed R, Salamanca C, Li X, Xu J, Kumar D, Rosen EM, Saha T. BRCA1 regulates follistatin function in ovarian cancer and human ovarian surface epithelial cells. PLoS One 2012; 7:e37697. [PMID: 22685544 PMCID: PMC3365892 DOI: 10.1371/journal.pone.0037697] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 04/26/2012] [Indexed: 11/19/2022] Open
Abstract
Follistatin (FST), a folliculogenesis regulating protein, is found in relatively high concentrations in female ovarian tissues. FST acts as an antagonist to Activin, which is often elevated in human ovarian carcinoma, and thus may serve as a potential target for therapeutic intervention against ovarian cancer. The breast cancer susceptibility gene 1 (BRCA1) is a known tumor suppressor gene in human breast cancer; however its role in ovarian cancer is not well understood. We performed microarray analysis on human ovarian carcinoma cell line SKOV3 that stably overexpress wild-type BRCA1 and compared with the corresponding empty vector-transfected clones. We found that stable expression of BRCA1 not only stimulates FST secretion but also simultaneously inhibits Activin expression. To determine the physiological importance of this phenomenon, we further investigated the effect of cellular BRCA1 on the FST secretion in immortalized ovarian surface epithelial (IOSE) cells derived from either normal human ovaries or ovaries of an ovarian cancer patient carrying a mutation in BRCA1 gene. Knock-down of BRCA1 in normal IOSE cells demonstrates down-regulation of FST secretion along with the simultaneous up-regulation of Activin expression. Furthermore, knock-down of FST in IOSE cell lines as well as SKOV3 cell line showed significantly reduced cell proliferation and decreased cell migration when compared with the respective controls. Thus, these findings suggest a novel function for BRCA1 as a regulator of FST expression and function in human ovarian cells.
Collapse
Affiliation(s)
- Tejaswita M. Karve
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C., United States of America
- Department of Biochemistry and Cellular and Molecular Biology, Georgetown University School of Medicine, Washington, D.C., United States of America
| | - Anju Preet
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Rosie Sneed
- University of District of Columbia, Washington, D.C., United States of America
| | - Clara Salamanca
- Canadian Ovarian Tissue Bank, BC Cancer Research Centre, Vancouver, B.C., Canada
| | - Xin Li
- Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University School of Medicine, Washington, D.C., United States of America
| | - Jingwen Xu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Deepak Kumar
- University of District of Columbia, Washington, D.C., United States of America
| | - Eliot M. Rosen
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Tapas Saha
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C., United States of America
- * E-mail: (TS)
| |
Collapse
|
22
|
Xia W, Fu W, Cai L, Kong H, Cai X, Liu J, Wang Y, Zou M, Xu D. Identification and characterization of FHL3 as a novel angiogenin-binding partner. Gene 2012; 504:233-7. [PMID: 22633874 DOI: 10.1016/j.gene.2012.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 05/02/2012] [Accepted: 05/10/2012] [Indexed: 12/14/2022]
Abstract
Angiogenin (Ang) is known to induce cell proliferation and inhibit apoptosis by cellular signaling pathways and by direct nuclear functions of Ang, but the mechanism of action for Ang is not yet clear. The aim of present study was to identify novel binding partner of Ang and to explore the underlying mechanism. With the use of yeast two-hybrid screening system, Ang was used as the bait to screen human fetal hepatic cDNA library for interacting proteins. Four and a half LIM domains 3 (FHL3) was identified as a novel Ang binding partner. The interaction between Ang and the full length FHL3 was further confirmed by yeast two-hybrid assay, co-immunoprecipitation and GST pull-down assays. Furthermore, FHL3 was required for Ang-mediated HeLa cell proliferation and nuclear translocation of Ang. These findings suggest that the interaction between Ang and FHL3 may provide some clues to the mechanisms of Ang-regulated cell growth and apoptosis.
Collapse
Affiliation(s)
- Wenrong Xia
- Laboratory of Genome Engineering, Beijing Institute of Basic Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Pyatibratov MG, Kostyukova AS. New insights into the role of angiogenin in actin polymerization. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 295:175-98. [PMID: 22449490 DOI: 10.1016/b978-0-12-394306-4.00011-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Angiogenin is a potent stimulator of angiogenesis. It interacts with endothelial cells and induces a wide range of cellular responses initiating a process of blood vessel formation. One important target of angiogenin is endothelial cell-surface actin, and their interaction might be one of crucial steps in angiogenin-induced neovascularization. Recently, it was shown that angiogenin inhibits polymerization of G-actin and changes the physical properties of F-actin. These observations suggest that angiogenin may cause changes in the cell cytoskeleton. This chapter reviews the current state of the literature regarding angiogenin structure and function and discusses the relationship between the angiogenin and actin and possible functional roles of their interaction.
Collapse
Affiliation(s)
- Mikhail G Pyatibratov
- Institute of Protein Research, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | | |
Collapse
|
24
|
Follistatin improves skeletal muscle healing after injury and disease through an interaction with muscle regeneration, angiogenesis, and fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:915-30. [PMID: 21689628 DOI: 10.1016/j.ajpath.2011.04.008] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Revised: 03/25/2011] [Accepted: 04/05/2011] [Indexed: 01/05/2023]
Abstract
Recovery from skeletal muscle injury is often incomplete because of the formation of fibrosis and inadequate myofiber regeneration; therefore, injured muscle could benefit significantly from therapies that both stimulate muscle regeneration and inhibit fibrosis. To this end, we focused on blocking myostatin, a member of the transforming growth factor-β superfamily and a negative regulator of muscle regeneration, with the myostatin antagonist follistatin. In vivo, follistatin-overexpressing transgenic mice underwent significantly greater myofiber regeneration and had less fibrosis formation compared with wild-type mice after skeletal muscle injury. Follistatin's mode of action is likely due to its ability to block myostatin and enhance neovacularization. Furthermore, muscle progenitor cells isolated from follistatin-overexpressing mice were significantly superior to muscle progenitors isolated from wild-type mice at regenerating dystrophin-positive myofibers when transplanted into the skeletal muscle of dystrophic mdx/severe combined immunodeficiency mice. In vitro, follistatin stimulated myoblasts to express MyoD, Myf5, and myogenin, which are myogenic transcription factors that promote myogenic differentiation. Moreover, follistatin's ability to enhance muscle differentiation is at least partially due to its ability to block myostatin, activin A, and transforming growth factor-β1, all of which are negative regulators of muscle cell differentiation. The findings of this study suggest that follistatin is a promising agent for improving skeletal muscle healing after injury and muscle diseases, such as the muscular dystrophies.
Collapse
|
25
|
George AL, Bangalore-Prakash P, Rajoria S, Suriano R, Shanmugam A, Mittelman A, Tiwari RK. Endothelial progenitor cell biology in disease and tissue regeneration. J Hematol Oncol 2011; 4:24. [PMID: 21609465 PMCID: PMC3123653 DOI: 10.1186/1756-8722-4-24] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 05/24/2011] [Indexed: 12/20/2022] Open
Abstract
Endothelial progenitor cells are increasingly being studied in various diseases ranging from ischemia, diabetic retinopathy, and in cancer. The discovery that these cells can be mobilized from their bone marrow niche to sites of inflammation and tumor to induce neovasculogenesis has afforded a novel opportunity to understand the tissue microenvironment and specific cell-cell interactive pathways. This review provides a comprehensive up-to-date understanding of the physiological function and therapeutic utility of these cells. The emphasis is on the systemic factors that modulate their differentiation/mobilization and survival and presents the challenges of its potential therapeutic clinical utility as a diagnostic and prognostic reagent.
Collapse
Affiliation(s)
- Andrea L George
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Gao X, Wei S, Lai K, Sheng J, Su J, Zhu J, Dong H, Hu H, Xu Z. Nucleolar follistatin promotes cancer cell survival under glucose-deprived conditions through inhibiting cellular rRNA synthesis. J Biol Chem 2010; 285:36857-64. [PMID: 20843798 DOI: 10.1074/jbc.m110.144477] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Solid tumor development is frequently accompanied by energy-deficient conditions such as glucose deprivation and hypoxia. Follistatin (FST), a secretory protein originally identified from ovarian follicular fluid, has been suggested to be involved in tumor development. However, whether it plays a role in cancer cell survival under energy-deprived conditions remains elusive. In this study, we demonstrated that glucose deprivation markedly enhanced the expression and nucleolar localization of FST in HeLa cells. The nucleolar localization of FST relied on its nuclear localization signal (NLS) comprising the residues 64-87. Localization of FST to the nucleolus attenuated rRNA synthesis, a key process for cellular energy homeostasis and cell survival. Overexpression of FST delayed glucose deprivation-induced apoptosis, whereas down-regulation of FST exerted the opposite effect. These functions depended on the presence of an intact NLS because the NLS-deleted mutant of FST lost the rRNA inhibition effect and the cell protective effect. Altogether, we identified a novel nucleolar function of FST, which is of importance in the modulation of cancer cell survival in response to glucose deprivation.
Collapse
Affiliation(s)
- Xiangwei Gao
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Serum follistatin in patients with prostate cancer metastatic to the bone. Clin Exp Metastasis 2010; 27:549-55. [PMID: 20623366 DOI: 10.1007/s10585-010-9344-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 07/01/2010] [Indexed: 01/19/2023]
Abstract
The clinical significance of circulating follistatin (FLST), an inhibitor of the multifunctional cytokine activin A (Act A), was investigated in patients with prostate cancer (PCa). The serum concentrations of this molecule were determined by an enzyme-linked immunosorbent assay (ELISA) in PCa patients with (M+) or without (M0) bone metastases, in patients with benign prostate hyperplasia (BPH) and in healthy subjects (HS). The effectiveness of FLST in detecting PCa patients with skeletal metastases was determined by the receiver operating characteristic (ROC) curve analysis. Serum FLST was significantly higher in PCa patients than in BPH patients (P = 0.001) or HS (P = 0.011). Conversely, in BPH patients, FLST levels resulted lower than in HS (P = 0.025). In cancer patients the serum concentrations of FLST significantly correlated with the presence of bone metastases (P = 0.0005) or increased PSA levels (P = 0.04). Interestingly, significant differences in the ratio between FLST and Act A serum concentrations (FLST/Act A) were observed between HS and BPH patients (P = 0.001) or PCa patients (P = 0.0005). Finally, ROC curve analysis, highlighted a sound diagnostic performance of FLST in detecting M+ patients (P = 0.0001). However, the diagnostic effectiveness of FLST did not result significantly superior to that of Act A or PSA. These findings suggest that FLST may be regarded as a potential, molecular target in the treatment of metastatic bone disease while its clinical role as soluble marker in the clinical management of PCa patients with bone metastases needs to be better defined.
Collapse
|
28
|
Binding of human angiogenin inhibits actin polymerization. Arch Biochem Biophys 2010; 495:74-81. [PMID: 20045391 DOI: 10.1016/j.abb.2009.12.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Revised: 12/25/2009] [Accepted: 12/29/2009] [Indexed: 11/22/2022]
Abstract
Angiogenin is a potent inducer of angiogenesis, a process of blood vessel formation. It interacts with endothelial and other cells and elicits a wide range of cellular responses including migration, proliferation, and tube formation. One important target of angiogenin is endothelial cell-surface actin and their interaction might be one of essential steps in angiogenin-induced neovascularization. Based on earlier indications that angiogenin promotes actin polymerization, we studied the binding interactions between angiogenin and actin in a wide range of conditions. We showed that at subphysiological KCl concentrations, angiogenin does not promote, but instead inhibits polymerization by sequestering G-actin. At low KCl concentrations angiogenin induces formation of unstructured aggregates, which, as shown by NMR, may be caused by angiogenin's propensity to form oligomers. Binding of angiogenin to preformed F-actin does not cause depolymerization of actin filaments though it causes their stiffening. Binding of tropomyosin and angiogenin to F-actin is not competitive at concentrations sufficient for saturation of actin filaments. These observations suggest that angiogenin may cause changes in the cell cytoskeleton by inhibiting polymerization of G-actin and changing the physical properties of F-actin.
Collapse
|
29
|
Kreidl E, Oztürk D, Metzner T, Berger W, Grusch M. Activins and follistatins: Emerging roles in liver physiology and cancer. World J Hepatol 2009; 1:17-27. [PMID: 21160961 PMCID: PMC2999257 DOI: 10.4254/wjh.v1.i1.17] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Revised: 09/10/2009] [Accepted: 09/17/2009] [Indexed: 02/06/2023] Open
Abstract
Activins are secreted proteins belonging to the TGF-β family of signaling molecules. Activin signals are crucial for differentiation and regulation of cell proliferation and apoptosis in multiple tissues. Signal transduction by activins relies mainly on the Smad pathway, although the importance of crosstalk with additional pathways is increasingly being recognized. Activin signals are kept in balance by antagonists at multiple levels of the signaling cascade. Among these, follistatin and FLRG, two members of the emerging family of follistatin-like proteins, can bind secreted activins with high affinity, thereby blocking their access to cell surface-anchored activin receptors. In the liver, activin A is a major negative regulator of hepatocyte proliferation and can induce apoptosis. The functions of other activins expressed by hepatocytes have yet to be more clearly defined. Deregulated expression of activins and follistatin has been implicated in hepatic diseases including inflammation, fibrosis, liver failure and primary cancer. In particular, increased follistatin levels have been found in the circulation and in the tumor tissue of patients suffering from hepatocellular carcinoma as well as in animal models of liver cancer. It has been argued that up-regulation of follistatin protects neoplastic hepatocytes from activin-mediated growth inhibition and apoptosis. The use of follistatin as biomarker for liver tumor development is impeded, however, due to the presence of elevated follistatin levels already during preceding stages of liver disease. The current article summarizes our evolving understanding of the multi-faceted activities of activins and follistatins in liver physiology and cancer.
Collapse
Affiliation(s)
- Emanuel Kreidl
- Emanuel Kreidl, Deniz Öztürk, Thomas Metzner, Walter Berger, Michael Grusch, Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, Vienna A-1090, Austria
| | | | | | | | | |
Collapse
|
30
|
Kim WC, Lee CH. The role of mammalian ribonucleases (RNases) in cancer. Biochim Biophys Acta Rev Cancer 2009; 1796:99-113. [PMID: 19463900 DOI: 10.1016/j.bbcan.2009.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Revised: 05/08/2009] [Accepted: 05/13/2009] [Indexed: 01/01/2023]
Abstract
Ribonucleases (RNases) are a group of enzymes that cleave RNAs at phosphodiester bonds resulting in remarkably diverse biological consequences. This review focuses on mammalian RNases that are capable of, or potentially capable of, cleaving messenger RNA (mRNA) as well as other RNAs in cells and play roles in the development of human cancers. The aims of this review are to provide an overview of the roles of currently known mammalian RNases, and the evidence that associate them as regulators of tumor development. The roles of these RNases as oncoproteins and/or tumor suppressors in influencing cell growth, apoptosis, angiogenesis, and other cellular hallmarks of cancer will be presented and discussed. The RNases under discussion include RNases from the conventional mRNA decay pathways, RNases that are activated under cellular stress, RNases from the miRNA pathway, and RNases with multifunctional activity.
Collapse
Affiliation(s)
- Wan-Cheol Kim
- Chemistry Program, University of Northern British Columbia, 3333 University Way, Prince George, BC, Canada V2N 4Z9
| | | |
Collapse
|
31
|
Zhou N, Fan W, Li M. Angiogenin is expressed in human dermal papilla cells and stimulates hair growth. Arch Dermatol Res 2008; 301:139-49. [PMID: 18936943 DOI: 10.1007/s00403-008-0907-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Revised: 09/10/2008] [Accepted: 09/23/2008] [Indexed: 12/01/2022]
Abstract
The perifollicular vasculature undergoes hair-cycle dependent expansion and degeneration. Multiple soluble factors derived from dermal papilla cells (DPCs) may act on surrounding blood vessels to influence angiogenesis, growth and differentiation, and thereby regulate cyclic hair growth. The goal of this study was to examine the expression of angiogenin, a potent angiogenic factor, in human DPCs, and to determine its role in hair growth. Reverse transcription polymerase chain reaction (RT-PCR), western blotting, immunofluorescence and ELISA analyzes were used to investigate the expression of angiogenin in human DPCs, while semi-quantitative RT-PCR was used to assess angiogenin mRNA expression in murine skin phased at different stages of the hair cycle. We detected angiogenin expression in DPCs, where it was found to be localized to the cytoplasm. Angiogenin mRNA was expressed in murine skin in a hair-cycle dependent manner, with maximum levels observed at the late anagen. Local injection of angiogenin promoted skin angiogenesis and induced anagen VI. In vitro studies showed that angiogenin significantly enhanced the elongation of hair follicles, and stimulated DPCs and ORS keratinocytes to proliferate. Taken together, these findings show that angiogenin is expressed in human DPCs, where it might contribute to hair growth directly, by stimulating DPCs and ORS keratinocytes to proliferate, or indirectly, by inducing local vascularization.
Collapse
Affiliation(s)
- Naihui Zhou
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, 210029 Nanjing, Jiangsu, China
| | | | | |
Collapse
|
32
|
Abstract
Angiogenin induces angiogenesis by activating vessel endothelial and smooth muscle cells and triggering a number of biological processes, including cell migration, invasion, proliferation, and formation of tubular structures. It has been reported that angiogenin plays its functions mainly through four pathways: (1) exerting its ribonucleolytic activity; (2) binding to membrane actin and then inducing basement membrane degradation; (3) binding to a putative 170-kDa protein and subsequently transducing signal into cytoplasm; and (4) translocating into the nucleus of target cells directly and then enhancing ribosomal RNA transcription. Angiogenin can also translocate into the nucleus of cancer cells and induces the corresponding cell proliferation. Furthermore, angiogenin has neuroprotective activities in the central nervous system and the loss of its function may be related to amyotrophic lateral sclerosis. This review intends to conclude the mechanisms underlying these actions of angiogenin and give a perspective on future research.
Collapse
Affiliation(s)
- Xiangwei Gao
- Research Center for Environmental Genomics, Bioelectromagnetics Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
| | | |
Collapse
|