1
|
Umetsu A, Watanabe M, Sato T, Higashide M, Nishikiori N, Furuhashi M, Ohguro H. TGF-β effects on adipogenesis of 3T3-L1 cells differ in 2D and 3D cell culture conditions. FEBS Open Bio 2024. [PMID: 39380256 DOI: 10.1002/2211-5463.13890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/04/2024] [Accepted: 08/23/2024] [Indexed: 10/10/2024] Open
Abstract
The TGF-β superfamily plays a pivotal role in the regulation of adipogenesis, but little is known about the potential differential role of the three isoforms of TGF-β, TGF-β-1~3. To further elucidate their role, two-dimensionally (2D) and three-dimensionally (3D) cultured 3T3-L1 mouse preadipocytes were subjected to the following analyses: (a) qPCR analysis of adipogenesis-related factors and major extracellular matrix protein (2D and /or 3D), (b) lipid staining by Oil Red O (2D) or BODIPY (3D), (c) Seahorse cellular metabolic measurement (2D), and (d) size and stiffness measurements of 3D 3T3-L1 spheroids. In the 2D cultured 3T3-L1 cells, mRNA expression levels of adipogenesis-related genes and Oil Red O lipid staining intensity were significantly increased by adipogenesis and they were substantially decreased following treatment with 0.1 nm TGF-β isoforms, with TGF-β2 having the greater effects. Consistent with these results, treatment with TGF-β2 resulted in suppression of mitochondrial and glycolytic functions in 2D cultured 3T3-L1 cells. However, the inhibitory effect of TGF-β on adipogenesis decreased under 3D spheroid culture conditions and TGF-β isoforms did not affect adipogenesis-induced (a) enlargement and downsizing of 3T3-L1 spheroids, (b) increase in BODIPY lipid staining intensity, and (c) up-regulation of the mRNA expression of adipogenesis-related genes. The findings presented herein suggest that the three TGF-β isoforms have different suppressive effects on adipogenesis-related cellular properties of 2D cultured 3T3-L1 cells and that their effects decrease under 3D spheroid culture conditions.
Collapse
Affiliation(s)
- Araya Umetsu
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Megumi Watanabe
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Japan
| | - Megumi Higashide
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Nami Nishikiori
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Japan
| | - Hiroshi Ohguro
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| |
Collapse
|
2
|
English J, Orofino J, Cederquist CT, Paul I, Li H, Auwerx J, Emili A, Belkina A, Cardamone D, Perissi V. GPS2-mediated regulation of the adipocyte secretome modulates adipose tissue remodeling at the onset of diet-induced obesity. Mol Metab 2023; 69:101682. [PMID: 36731652 PMCID: PMC9922684 DOI: 10.1016/j.molmet.2023.101682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/22/2023] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE Dysfunctional, unhealthy expansion of white adipose tissue due to excess dietary intake is a process at the root of obesity and Type 2 Diabetes development. The objective of this study is to contribute to a better understanding of the underlying mechanism(s) regulating the early stages of adipose tissue expansion and adaptation to dietary stress due to an acute, high-fat diet (HFD) challenge, with a focus on the communication between adipocytes and other stromal cells. METHODS We profiled the early response to high-fat diet exposure in wildtype and adipocyte-specific GPS2-KO (GPS2-AKO) mice at the cellular, tissue and organismal level. A multi-pronged approach was employed to disentangle the complex cellular interactions dictating tissue remodeling, via single-cell RNA sequencing and FACS profiling of the stromal fraction, and semi-quantitative proteomics of the adipocyte-derived exosomal cargo after 5 weeks of HFD feeding. RESULTS Our results indicate that loss of GPS2 in mature adipocytes leads to impaired adaptation to the metabolic stress imposed by HFD feeding. GPS2-AKO mice are significantly more inflamed, insulin resistant, and obese, compared to the WT counterparts. At the cellular level, lack of GPS2 in adipocytes impacts upon other stromal populations, with both the eWAT and scWAT depots exhibiting changes in the immune and non-immune compartments that contribute to an increase in inflammatory and anti-adipogenic cell types. Our studies also revealed that adipocyte to stromal cell communication is facilitated by exosomes, and that transcriptional rewiring of the exosomal cargo is crucial for tissue remodeling. Loss of GPS2 results in increased expression of secreted factors promoting a TGFβ-driven fibrotic microenvironment favoring unhealthy tissue remodeling and expansion. CONCLUSIONS Adipocytes serve as an intercellular signaling hub, communicating with the stromal compartment via paracrine signaling. Our study highlights the importance of proper regulation of the 'secretome' released by energetically stressed adipocytes at the onset of obesity. Altered transcriptional regulation of factors secreted via adipocyte-derived exosomes (AdExos), in the absence of GPS2, contributes to the establishment of an anti-adipogenic, pro-fibrotic adipose tissue environment, and to hastened progression towards a metabolically dysfunctional phenotype.
Collapse
Affiliation(s)
- Justin English
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Joseph Orofino
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.
| | - Carly T. Cederquist
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Indranil Paul
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; Center for Network Systems Biology, Boston University, Boston, MA, USA.
| | - Hao Li
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland.
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland.
| | - Andrew Emili
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; Center for Network Systems Biology, Boston University, Boston, MA, USA.
| | - Anna Belkina
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA.
| | - Dafne Cardamone
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA.
| | - Valentina Perissi
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA; School of Life Science, Northwestern Polytechnical University, Xi'an 710072, China.
| |
Collapse
|
3
|
Huang CJ, Choo KB, Chen CF. The MicroRNA-Signaling-Peroxisome Proliferator-Activated Receptor Gamma Connection in the Modulation of Adipogenesis: Bioinformatics Projection on Chicken. Poult Sci 2022; 101:101950. [PMID: 35689996 PMCID: PMC9192975 DOI: 10.1016/j.psj.2022.101950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 03/19/2022] [Accepted: 04/15/2022] [Indexed: 10/29/2022] Open
|
4
|
Balazova L, Balaz M, Horvath C, Horváth Á, Moser C, Kovanicova Z, Ghosh A, Ghoshdastider U, Efthymiou V, Kiehlmann E, Sun W, Dong H, Ding L, Amri EZ, Nuutila P, Virtanen KA, Niemi T, Ukropcova B, Ukropec J, Pelczar P, Lamla T, Hamilton B, Neubauer H, Wolfrum C. GPR180 is a component of TGFβ signalling that promotes thermogenic adipocyte function and mediates the metabolic effects of the adipocyte-secreted factor CTHRC1. Nat Commun 2021; 12:7144. [PMID: 34880217 PMCID: PMC8655035 DOI: 10.1038/s41467-021-27442-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022] Open
Abstract
Activation of thermogenic brown and beige adipocytes is considered as a strategy to improve metabolic control. Here, we identify GPR180 as a receptor regulating brown and beige adipocyte function and whole-body glucose homeostasis, whose expression in humans is associated with improved metabolic control. We demonstrate that GPR180 is not a GPCR but a component of the TGFβ signalling pathway and regulates the activity of the TGFβ receptor complex through SMAD3 phosphorylation. In addition, using genetic and pharmacological tools, we provide evidence that GPR180 is required to manifest Collagen triple helix repeat containing 1 (CTHRC1) action to regulate brown and beige adipocyte activity and glucose homeostasis. In this work, we show that CTHRC1/GPR180 signalling integrates into the TGFβ signalling as an alternative axis to fine-tune and achieve low-grade activation of the pathway to prevent pathophysiological response while contributing to control of glucose and energy metabolism.
Collapse
Affiliation(s)
- Lucia Balazova
- Institute of Food, Nutrition and Health, ETH Zürich, 8603, Schwerzenbach, Switzerland
| | - Miroslav Balaz
- Institute of Food, Nutrition and Health, ETH Zürich, 8603, Schwerzenbach, Switzerland
- Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, 84505, Bratislava, Slovakia
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, 84215, Bratislava, Slovakia
| | - Carla Horvath
- Institute of Food, Nutrition and Health, ETH Zürich, 8603, Schwerzenbach, Switzerland
| | - Áron Horváth
- Biomechanics Laboratory, University Hospital Balgrist, University of Zurich, 8008, Zurich, Switzerland
- Institute of Biomechanics, ETH Zurich, 8093, Zurich, Switzerland
| | - Caroline Moser
- Institute of Food, Nutrition and Health, ETH Zürich, 8603, Schwerzenbach, Switzerland
| | - Zuzana Kovanicova
- Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, 84505, Bratislava, Slovakia
| | - Adhideb Ghosh
- Institute of Food, Nutrition and Health, ETH Zürich, 8603, Schwerzenbach, Switzerland
- Functional Genomics Centre Zurich, ETH Zurich/ University of Zurich, 8057, Zurich, Switzerland
| | - Umesh Ghoshdastider
- Institute of Food, Nutrition and Health, ETH Zürich, 8603, Schwerzenbach, Switzerland
| | - Vissarion Efthymiou
- Institute of Food, Nutrition and Health, ETH Zürich, 8603, Schwerzenbach, Switzerland
| | - Elke Kiehlmann
- Institute of Food, Nutrition and Health, ETH Zürich, 8603, Schwerzenbach, Switzerland
| | - Wenfei Sun
- Institute of Food, Nutrition and Health, ETH Zürich, 8603, Schwerzenbach, Switzerland
| | - Hua Dong
- Institute of Food, Nutrition and Health, ETH Zürich, 8603, Schwerzenbach, Switzerland
| | - Lianggong Ding
- Institute of Food, Nutrition and Health, ETH Zürich, 8603, Schwerzenbach, Switzerland
| | - Ez-Zoubir Amri
- Université Côte d'Azur, French National Centre for Scientific Research, Inserm, iBV, 06107, Nice, France
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, 20520, Turku, Finland
| | | | - Tarja Niemi
- Department of Surgery, Turku University Hospital, 20520, Turku, Finland
| | - Barbara Ukropcova
- Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, 84505, Bratislava, Slovakia
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, 81108, Bratislava, Slovakia
| | - Jozef Ukropec
- Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, 84505, Bratislava, Slovakia
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, 3350, Basel, Switzerland
| | - Thorsten Lamla
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397, Biberach an der Riss, Germany
| | - Bradford Hamilton
- Cardiometabolic Diseases Research Department, Boehringer Ingelheim Pharma GmbH and Co. KG, 88397, Biberach an der Riss, Germany
| | - Heike Neubauer
- Cardiometabolic Diseases Research Department, Boehringer Ingelheim Pharma GmbH and Co. KG, 88397, Biberach an der Riss, Germany
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zürich, 8603, Schwerzenbach, Switzerland.
| |
Collapse
|
5
|
Piccotti F, Rybinska I, Scoccia E, Morasso C, Ricciardi A, Signati L, Triulzi T, Corsi F, Truffi M. Lipofilling in Breast Oncological Surgery: A Safe Opportunity or Risk for Cancer Recurrence? Int J Mol Sci 2021; 22:ijms22073737. [PMID: 33916703 PMCID: PMC8038405 DOI: 10.3390/ijms22073737] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 02/08/2023] Open
Abstract
Lipofilling (LF) is a largely employed technique in reconstructive and esthetic breast surgery. Over the years, it has demonstrated to be extremely useful for treatment of soft tissue defects after demolitive or conservative breast cancer surgery and different procedures have been developed to improve the survival of transplanted fat graft. The regenerative potential of LF is attributed to the multipotent stem cells found in large quantity in adipose tissue. However, a growing body of pre-clinical evidence shows that adipocytes and adipose-derived stromal cells may have pro-tumorigenic potential. Despite no clear indication from clinical studies has demonstrated an increased risk of cancer recurrence upon LF, these observations challenge the oncologic safety of the procedure. This review aims to provide an updated overview of both the clinical and the pre-clinical indications to the suitability and safety of LF in breast oncological surgery. Cellular and molecular players in the crosstalk between adipose tissue and cancer are described, and heterogeneous contradictory results are discussed, highlighting that important issues still remain to be solved to get a clear understanding of LF safety in breast cancer patients.
Collapse
Affiliation(s)
- Francesca Piccotti
- Laboratorio di Nanomedicina ed Imaging Molecolare, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (F.P.); (C.M.); (A.R.)
| | - Ilona Rybinska
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (I.R.); (T.T.)
| | - Elisabetta Scoccia
- Breast Unit, Surgery Department, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (E.S.); (F.C.)
| | - Carlo Morasso
- Laboratorio di Nanomedicina ed Imaging Molecolare, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (F.P.); (C.M.); (A.R.)
| | - Alessandra Ricciardi
- Laboratorio di Nanomedicina ed Imaging Molecolare, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (F.P.); (C.M.); (A.R.)
| | - Lorena Signati
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università Degli Studi di Milano, 20157 Milano, Italy;
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (I.R.); (T.T.)
| | - Fabio Corsi
- Breast Unit, Surgery Department, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (E.S.); (F.C.)
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università Degli Studi di Milano, 20157 Milano, Italy;
| | - Marta Truffi
- Laboratorio di Nanomedicina ed Imaging Molecolare, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (F.P.); (C.M.); (A.R.)
- Correspondence: ; Tel.: +39-0382-592219
| |
Collapse
|
6
|
Bielczyk-Maczynska E. White Adipocyte Plasticity in Physiology and Disease. Cells 2019; 8:E1507. [PMID: 31775295 PMCID: PMC6953026 DOI: 10.3390/cells8121507] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 12/15/2022] Open
Abstract
Cellular plasticity is a transformation of a terminally differentiated cell into another cell type, which has been long known to occur in disease and regeneration. However, white adipocytes (fat cells) have only recently been observed to undergo different types of cellular plasticity. Adipocyte transdifferentiation into myofibroblasts and cancer-associated fibroblasts occurs in fibrosis and cancer, respectively. On the other hand, reversible adipocyte dedifferentiation into adipocyte progenitor cells (preadipocytes) has been demonstrated in mammary gland and in dermal adipose tissue. Here we discuss the research on adipocyte plasticity, including the experimental approaches that allowed to detect and study it, the current state of the knowledge, major research questions which remain to be addressed, and the advances required to stimulate adipocyte plasticity research. In the future, the knowledge of the molecular mechanisms of adipocyte plasticity can be utilized both to prevent adipocyte plasticity in disease and to stimulate it for use in regenerative medicine.
Collapse
Affiliation(s)
- Ewa Bielczyk-Maczynska
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
7
|
Volz AC, Omengo B, Gehrke S, Kluger PJ. Comparing the use of differentiated adipose-derived stem cells and mature adipocytes to model adipose tissue in vitro. Differentiation 2019; 110:19-28. [PMID: 31568881 DOI: 10.1016/j.diff.2019.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/19/2019] [Accepted: 09/03/2019] [Indexed: 12/14/2022]
Abstract
In vitro models of human adipose tissue may serve as beneficial alternatives to animal models to study basic biological processes, identify new drug targets, and as soft tissue implants. With this approach, we aimed to evaluate adipose-derived stem cells (ASC) and mature adipocytes (MA) comparatively for the application in the in vitro setup of adipose tissue constructs to imitate native adipose tissue physiology. We used human primary MAs and human ASCs, differentiated for 14 days, and encapsulated them in collagen type I hydrogels to build up a three-dimensional (3D) adipose tissue model. The maintenance of the models was analyzed after seven days based on a viability staining. Further, the expression of the adipocyte specific protein perilipin A and the release of leptin and glycerol were evaluated. Gene transcription profiles of models based on dASCs and MAs were analyzed with regard to native adipose tissue. Compared to MAs, dASCs showed an immature differentiation state. Further, gene transcription of MAs suggests a behavior closer to native tissue in terms of angiogenesis, which supports MAs as preferred cell type. In contrast to native adipose tissue, genes of de novo lipogenesis and tissue remodeling were upregulated in the in vitro attempts.
Collapse
Affiliation(s)
- Ann-Cathrin Volz
- Reutlingen Research Institute, Reutlingen University, Alteburgstrasse 150, 72762, Reutlingen, Germany; University of Hohenheim, Schloss Hohenheim 1, 70599, Stuttgart, Germany
| | - Birgit Omengo
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstrasse 12, 70569, Stuttgart, Germany
| | - Sandra Gehrke
- Research & Development, Research Special Skincare, Beiersdorf AG, Unnastrasse 48, 20253, Hamburg, Germany
| | - Petra Juliane Kluger
- Reutlingen Research Institute, Reutlingen University, Alteburgstrasse 150, 72762, Reutlingen, Germany; Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Cell and Tissue Engineering, Nobelstrasse 12, 70569, Stuttgart, Germany.
| |
Collapse
|
8
|
Tan J, Wang Y, Wang S, Wu S, Yuan Z, Zhu X. Label-free quantitative proteomics identifies transforming growth factor β1 (TGF-β1) as an inhibitor of adipogenic transformation in OP9-DL1 cells and primary thymic stromal cells. Cell Biosci 2019; 9:48. [PMID: 31249661 PMCID: PMC6570845 DOI: 10.1186/s13578-019-0311-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/08/2019] [Indexed: 11/21/2022] Open
Abstract
Background Adipocyte accumulation is a predominant feature of age-related thymic involution, but the mechanisms responsible for thymic adipogenesis remain to be elucidated. The aim of this study was to identify key regulators in thymic adipogenesis. We used rosiglitazone, a potent peroxisome proliferator-activated receptor γ (PPARγ) agonist, to induce adipogenic differentiation of OP9-DL1 cells, and investigated the differentially expressed proteins during adipogenic differentiation by using label-free quantitative proteomics. Furthermore, the effects of transforming growth factor β1 (TGF-β1) on rosiglitazone-induced adipogenic differentiation of OP9-DL1 cells as well as the underlying mechanisms were also investigated. Results Proteomic analysis identified 139 proteins differed significantly in rosiglitazone-treated cells compared with dimethyl sulphoxide (DMSO)-treated cells. Rosiglitazone-induced adipogenic differentiation was inhibited by TGF-β1 treatment in OP9-DL1 cells and primary thymic stromal cells. Real-time PCR analysis showed significant increases in PPARγ and fatty acid binding protein 4 mRNA levels in rosiglitazone-treated cells, which were inhibited by TGF-β1 treatment. TGF-β1 down-regulated PPARγ expression at both mRNA and protein levels in OP9-DL1 cells. Chromatin immunoprecipitation analysis demonstrated that TGF-β1 enhanced the binding of Smad2/3 and histone deacetylase 1, but reduced the binding of H3K14ac to the promoter of PPARγ gene. TGF-β1 partially reversed the inhibitory effects of rosiglitazone on the expression of Axin2 and β-catenin protein levels. TGF-β1 inhibited rosiglitazone-induced adipogenic transformation in OP9-DL1 cells by down-regulation of PPARγ and activation of the canonical Wnt/β-catenin signaling pathway. Conclusion Taken together, activation of TGF-β pathway may serve as a useful strategy to prevent thymic adiposity in age-related thymic involution. Electronic supplementary material The online version of this article (10.1186/s13578-019-0311-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jianxin Tan
- 1State Key Laboratory of Reproductive Medicine, Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004 People's Republic of China
| | - Yajun Wang
- 2Research Center, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004 People's Republic of China
| | - Siliang Wang
- 3Department of Medical Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022 People's Republic of China
| | - Simeng Wu
- 4Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, 110022 People's Republic of China
| | - Zhe Yuan
- 4Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, 110022 People's Republic of China
| | - Xike Zhu
- 2Research Center, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004 People's Republic of China
| |
Collapse
|
9
|
Yoshioka H, Yoshiko Y. The Roles of Long Non-Protein-Coding RNAs in Osteo-Adipogenic Lineage Commitment. Int J Mol Sci 2017; 18:E1236. [PMID: 28598385 PMCID: PMC5486059 DOI: 10.3390/ijms18061236] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 12/17/2022] Open
Abstract
Osteoblasts and adipocytes share a common mesenchymal progenitor in the bone marrow. This implies that a reciprocal relationship exists between osteogenic and adipogenic differentiation. Further, cells of osteoblast lineage transdifferentiate into adipocytes under some circumstances. Dysregulation of osteo-adipogenic fate-determination leads to bone diseases such as osteoporosis, accompanied by an increase in bone marrow adipose tissue. Thus, the fine-tuning of osteo-adipogenesis is necessary for bone homeostasis. Osteo-adipogenic progression is governed by a complex crosstalk of extrinsic signals, transcription factors, and epigenetic factors. Long non-protein-coding RNAs (lncRNAs) act in part as epigenetic regulators in a broad range of biological activities, such as chromatin organization, transcriptional regulation, post-translational modifications, and histone modification. In this review, we highlight the roles of epigenetic regulators, particularly lncRNAs, in the osteo-adipogenic lineage commitment of bone marrow mesenchymal stem cells and the adipogenic transdifferentiation of osteoblasts.
Collapse
Affiliation(s)
- Hirotaka Yoshioka
- Department of Calcified Tissue Biology, Hiroshima University Institute of Biomedical and Health Sciences, 734-8553 Hiroshima, Japan.
| | - Yuji Yoshiko
- Department of Calcified Tissue Biology, Hiroshima University Institute of Biomedical and Health Sciences, 734-8553 Hiroshima, Japan.
| |
Collapse
|
10
|
EGF and hydrocortisone as critical factors for the co-culture of adipogenic differentiated ASCs and endothelial cells. Differentiation 2017; 95:21-30. [DOI: 10.1016/j.diff.2017.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 01/10/2017] [Accepted: 01/18/2017] [Indexed: 01/08/2023]
|
11
|
Curcio F, Ferro G, Basile C, Liguori I, Parrella P, Pirozzi F, Della-Morte D, Gargiulo G, Testa G, Tocchetti CG, Bonaduce D, Abete P. Biomarkers in sarcopenia: A multifactorial approach. Exp Gerontol 2016; 85:1-8. [DOI: 10.1016/j.exger.2016.09.007] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/05/2016] [Accepted: 09/08/2016] [Indexed: 12/11/2022]
|
12
|
Waked K, Colle J, Doornaert M, Cocquyt V, Blondeel P. Systematic review: The oncological safety of adipose fat transfer after breast cancer surgery. Breast 2016; 31:128-136. [PMID: 27837706 DOI: 10.1016/j.breast.2016.11.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 10/02/2016] [Accepted: 11/03/2016] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES Oncological concerns have risen around the safety of adipose fat transfer (AFT) after breast cancer surgery. In this article, we present the clinical and molecular evidences, and discuss the current contradiction between them. MATERIALS AND METHODS Every clinical trial and experimental study on AFT and its oncological influences was screened. Between September 2014 and September 2016, 856 articles from four databases were found. 105 core articles were selected. RESULTS A total of 18 clinical studies have been published. The loco-regional recurrence (LRR) incidence rates range between 0 and 3.90% per year. For the mastectomy and breast conservative therapy group separately, a LRR per year between 0 and 1.62% and 0-3.90 has been reported, respectively. Some studies included a matched control group and found no significant difference between cases and controls, with the exception of a subgroup of patients with intraepithelial breast carcinoma. Adipose derived mesenchymal stem cells have a potential oncogenic effect on residual cancer cells after breast cancer surgery. Numerous signalling proteins and pathways have been described that can stimulate tumour initiation and growth. CONCLUSION There is a contradiction between experimental and clinical findings. Numerous adipokines have been discovered that could potentially promote tumour initiation and growth, but clinical studies fail to point out a significant increase in LRR in patients who receive AFT after breast cancer surgery. More prospective studies are needed with a sufficient follow-up time and analysis of some critical factors, such as adjuvant radiotherapy and hormonal therapy, the origin and volume of the injected fat, and genetic influences.
Collapse
Affiliation(s)
- Karl Waked
- University Hospital of Ghent, De Pintelaan 185, 9000 Gent, Belgium.
| | - Julien Colle
- University Hospital of Ghent, De Pintelaan 185, 9000 Gent, Belgium.
| | - Maarten Doornaert
- Private Medical Center: Maaltebrugge Ghent, Maaltebruggestraat 288, 9000 Gent, Belgium.
| | | | - Phillip Blondeel
- University Hospital of Ghent, De Pintelaan 185, 9000 Gent, Belgium.
| |
Collapse
|
13
|
Grassot V, Bouchatal A, Da Silva A, Chantepie S, Papy-Garcia D, Maftah A, Gallet PF, Petit JM. Heparan sulfates and the decrease of N-glycans promote early adipogenic differentiation rather than myogenesis of murine myogenic progenitor cells. Differentiation 2016; 93:15-26. [PMID: 27689814 DOI: 10.1016/j.diff.2016.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 08/05/2016] [Accepted: 08/29/2016] [Indexed: 12/25/2022]
Abstract
In vitro, extracted muscle satellite cells, called myogenic progenitor cells, can differentiate either in myotubes or preadipocytes, depending on environmental factors and the medium. Transcriptomic analyses on glycosylation genes during satellite cells differentiation into myotubes showed that 31 genes present a significant variation of expression at the early stages of murine myogenic progenitor cells (MPC) differentiation. In the present study, we analyzed the expression of 383 glycosylation related genes during murine MPC differentiation into preadipocytes and compared the data to those previously obtained during their differentiation into myotubes. Fifty-six glycosylation related genes are specifically modified in their expression during early adipogenesis. The variations correspond mainly to: a decrease of N-glycans, and of alpha (2,3) and (2,6) linked sialic acids, and to a high level of heparan sulfates. A high amount of TGF-β1 in extracellular media during early adipogenesis was also observed. It seems that the increases of heparan sulfates and TGF-β1 favor pre-adipogenic differentition of MPC and possibly prevent their myogenic differentiation.
Collapse
Affiliation(s)
- Vincent Grassot
- INRA, UMR 1061, F-87060 Limoges, France; Université de Limoges, Faculté des Sciences et Techniques, Unité de Génétique Moléculaire Animale, UGMA, F-87060 Limoges, France.
| | - Amel Bouchatal
- INRA, UMR 1061, F-87060 Limoges, France; Université de Limoges, Faculté des Sciences et Techniques, Unité de Génétique Moléculaire Animale, UGMA, F-87060 Limoges, France.
| | - Anne Da Silva
- INRA, UMR 1061, F-87060 Limoges, France; Université de Limoges, Faculté des Sciences et Techniques, Unité de Génétique Moléculaire Animale, UGMA, F-87060 Limoges, France.
| | - Sandrine Chantepie
- CNRS, EAC 7149, F-94000 Créteil, France; Université Paris Est Créteil, Laboratoire Croissance, Régénération, Réparation et Régénération Tissulaires, CRRET, F-94000 Créteil, France.
| | - Dulce Papy-Garcia
- CNRS, EAC 7149, F-94000 Créteil, France; Université Paris Est Créteil, Laboratoire Croissance, Régénération, Réparation et Régénération Tissulaires, CRRET, F-94000 Créteil, France.
| | - Abderrahman Maftah
- INRA, UMR 1061, F-87060 Limoges, France; Université de Limoges, Faculté des Sciences et Techniques, Unité de Génétique Moléculaire Animale, UGMA, F-87060 Limoges, France.
| | - Paul-François Gallet
- INRA, UMR 1061, F-87060 Limoges, France; Université de Limoges, Faculté des Sciences et Techniques, Unité de Génétique Moléculaire Animale, UGMA, F-87060 Limoges, France.
| | - Jean-Michel Petit
- INRA, UMR 1061, F-87060 Limoges, France; Université de Limoges, Faculté des Sciences et Techniques, Unité de Génétique Moléculaire Animale, UGMA, F-87060 Limoges, France.
| |
Collapse
|
14
|
Combellack EJ, Jessop ZM, Naderi N, Griffin M, Dobbs T, Ibrahim A, Evans S, Burnell S, Doak SH, Whitaker IS. Adipose regeneration and implications for breast reconstruction: update and the future. Gland Surg 2016; 5:227-41. [PMID: 27047789 PMCID: PMC4791352 DOI: 10.3978/j.issn.2227-684x.2016.01.01] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/17/2015] [Indexed: 12/20/2022]
Abstract
The evolution of breast reconstruction and management of breast cancer has evolved significantly since the earliest descriptions in the Edwin Smith Papyrus (3,000 BC). The development of surgical and scientific expertise has changed the way that women are managed, and plastic surgeons are now able to offer a wide range of reconstructive options to suit individual needs. Beyond the gold standard autologous flap based reconstructions, regenerative therapies promise the elimination of donor site morbidity whilst providing equivalent aesthetic and functional outcomes. Future research aims to address questions regarding ideal cell source, optimisation of scaffold composition and interaction of de novo adipose tissue in the microenvironment of breast cancer.
Collapse
|
15
|
Moseti D, Regassa A, Kim WK. Molecular Regulation of Adipogenesis and Potential Anti-Adipogenic Bioactive Molecules. Int J Mol Sci 2016; 17:ijms17010124. [PMID: 26797605 PMCID: PMC4730365 DOI: 10.3390/ijms17010124] [Citation(s) in RCA: 529] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 12/27/2015] [Accepted: 01/07/2016] [Indexed: 11/24/2022] Open
Abstract
Adipogenesis is the process by which precursor stem cells differentiate into lipid laden adipocytes. Adipogenesis is regulated by a complex and highly orchestrated gene expression program. In mammalian cells, the peroxisome proliferator-activated receptor γ (PPARγ), and the CCAAT/enhancer binding proteins (C/EBPs) such as C/EBPα, β and δ are considered the key early regulators of adipogenesis, while fatty acid binding protein 4 (FABP4), adiponectin, and fatty acid synthase (FAS) are responsible for the formation of mature adipocytes. Excess accumulation of lipids in the adipose tissue leads to obesity, which is associated with cardiovascular diseases, type II diabetes and other pathologies. Thus, investigating adipose tissue development and the underlying molecular mechanisms is vital to develop therapeutic agents capable of curbing the increasing incidence of obesity and related pathologies. In this review, we address the process of adipogenic differentiation, key transcription factors and proteins involved, adipogenic regulators and potential anti-adipogenic bioactive molecules.
Collapse
Affiliation(s)
- Dorothy Moseti
- Department of Animal Science, University of Manitoba, 201 Animal Science building, Winnipeg, MB R3T 2N2, Canada.
| | - Alemu Regassa
- Department of Animal Science, University of Manitoba, 201 Animal Science building, Winnipeg, MB R3T 2N2, Canada.
| | - Woo-Kyun Kim
- Department of Poultry Science, University of Georgia, 303 Poultry Science Building, Athens, GA 30602-2772, USA.
| |
Collapse
|
16
|
Kalinkovich A, Livshits G. Sarcopenia--The search for emerging biomarkers. Ageing Res Rev 2015; 22:58-71. [PMID: 25962896 DOI: 10.1016/j.arr.2015.05.001] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 05/06/2015] [Accepted: 05/06/2015] [Indexed: 12/12/2022]
Abstract
Sarcopenia, an age-related decline in skeletal muscle mass and function, dramatically affects the life quality of elder people. In view of increasing life expectancy, sarcopenia renders a heavy burden on the health care system. However, although there is a consensus that sarcopenia is a multifactorial syndrome, its etiology, underlying mechanisms, and even definition remain poorly delineated, thus, preventing development of a precise treatment strategy. The main aim of our review is to critically analyze potential sarcopenia biomarkers in light of the molecular mechanisms of their involvement in sarcopenia pathogenesis. Normal muscle mass and function maintenance are proposed to be dependent on the dynamic balance between the positive regulators of muscle growth such as bone morphogenetic proteins (BMPs), brain-derived neurotrophic factor (BDNF), follistatin (FST) and irisin, and negative regulators including TGFβ, myostatin, activins A and B, and growth and differentiation factor-15 (GDF-15). We hypothesize that the shift in this balance to muscle growth inhibitors, along with increased expression of the C- terminal agrin fragment (CAF) associated with age-dependent neuromuscular junction (NMJ) dysfunction, as well as skeletal muscle-specific troponin T (sTnT), a key component of contractile machinery, is a main mechanism underlying sarcopenia pathogenesis. Thus, this review proposes and emphasizes that these molecules are the emerging sarcopenia biomarkers.
Collapse
|
17
|
Amable PR, Teixeira MVT, Carias RBV, Granjeiro JM, Borojevic R. Gene expression and protein secretion during human mesenchymal cell differentiation into adipogenic cells. BMC Cell Biol 2014; 15:46. [PMID: 25526965 PMCID: PMC4293810 DOI: 10.1186/s12860-014-0046-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 12/11/2014] [Indexed: 12/14/2022] Open
Abstract
Background Mesenchymal stromal cells (MSC) can be obtained from potentially any tissue from the human body, but cells purified from different sources are undoubtedly different, and for each medical application, the MSC with the best regenerative potential should be chosen. Results Bone marrow-derived mesenchymal stromal cells (BM-MSC), adipose tissue-derived mesenchymal stromal cells (AT-MSC) and Wharton’s Jelly-derived mesenchymal stromal cells (WJ-MSC) were isolated from human tissues and were cultured under differentiation media supplemented with fetal bovine serum. We quantified the expression of stem cell and adipocyte genetic markers using quantitative real time PCR, as well as the secretion of cytokines, extracellular matrix components and growth factors using Luminex and ELISA. All three MSC differentiated into adipogenic cells. AT-MSC showed the highest shift in ADIPOQ, CEBPA and PPARG mRNA expression. BM-MSC kept high expression levels of stem-cell markers SOX2 and POU5F1. WJ-MSC showed the lowest increase in mRNA expression when cells were induced to differentiate into adipocytes. Regarding protein secretion, adipocyte-like cells generated from WJ-MSC secreted the highest chemokine levels. AT-MSC-derived adipocyte-like cells secreted the lowest cytokine amounts and the highest quantity of collagen types I and III. Adipocyte-like cells obtained from BM-MSC secreted high amounts of most angiogenic factors, growth factors TGF-β1 and TGF-β2, collagens type II and IV, heparan sulfate, laminin and aggrecan. Conclusion Mesenchymal stromal cells purified from different tissues have a different behavior when induced to differentiate into adipocyte-like cells.
Collapse
Affiliation(s)
- Paola Romina Amable
- Excellion Biomedical Services S.A., Rua Afrânio de Mello Franco 333, Quitandinha, Petrópolis, Rio de Janeiro, Brazil.
| | | | | | - José Mauro Granjeiro
- National Institute of Metrology, Quality and Technology (Inmetro), Xerém, Rio de Janeiro, Brazil.
| | - Radovan Borojevic
- National Institute of Metrology, Quality and Technology (Inmetro), Xerém, Rio de Janeiro, Brazil. .,Faculdade de Medicina de Petrópolis, Faculdades Arthur Sá Earp Neto, Petrópolis, Rio de Janeiro, Brazil.
| |
Collapse
|
18
|
Vanderhoeven JP, Bierle CJ, Kapur RP, McAdams RM, Beyer RP, Bammler TK, Farin FM, Bansal A, Spencer M, Deng M, Gravett MG, Rubens CE, Rajagopal L, Adams Waldorf KM. Group B streptococcal infection of the choriodecidua induces dysfunction of the cytokeratin network in amniotic epithelium: a pathway to membrane weakening. PLoS Pathog 2014; 10:e1003920. [PMID: 24603861 PMCID: PMC3946355 DOI: 10.1371/journal.ppat.1003920] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 12/27/2013] [Indexed: 11/18/2022] Open
Abstract
Early events leading to intrauterine infection remain poorly defined, but may hold the key to preventing preterm delivery. To determine molecular pathways within fetal membranes (chorioamnion) associated with early choriodecidual infection that may progress to preterm premature rupture of membranes (PPROM), we examined the effects of a Group B Streptococcus (GBS) choriodecidual infection on chorioamnion in a nonhuman primate model. Ten chronically catheterized pregnant monkeys (Macaca nemestrina) at 118–125 days gestation (term = 172 days) received choriodecidual inoculation of either GBS (n = 5) or saline (n = 5). Cesarean section was performed in the first week after GBS or saline inoculation. RNA extracted from chorioamnion (inoculation site) was profiled by microarray. Single gene, Gene Set, and Ingenuity Pathway Analysis results were validated using qRT-PCR (chorioamnion), Luminex (amniotic fluid, AF), immunohistochemistry, and transmission electron microscopy (TEM). Despite uterine quiescence in most cases, significant elevations of AF cytokines (TNF-α, IL-8, IL-1β, IL-6) were detected in GBS versus controls (p<0.05). Choriodecidual infection resolved by the time of cesarean section in 3 of 5 cases and GBS was undetectable by culture and PCR in the AF. A total of 331 genes were differentially expressed (>2-fold change, p<0.05). Remarkably, GBS exposure was associated with significantly downregulated expression of multiple cytokeratin (CK) and other cytoskeletal genes critical for maintenance of tissue tensile strength. Immunofluorescence revealed highly significant changes in the CK network within amniocytes with dense CK aggregates and retraction from the cell periphery (all p = 0.006). In human pregnancies affected by PPROM, there was further evidence of CK network retraction with significantly shorter amniocyte foot processes (p = 0.002). These results suggest early choriodecidual infection results in decreased cellular membrane integrity and tensile strength via dysfunction of CK networks. Downregulation of CK expression and perturbations in the amniotic epithelial cell intermediate filament network occur after GBS choriodecidual infection, which may contribute to PPROM. Group B Streptococcus (GBS) is one cause of preterm birth, stillbirth, and fetal brain injury. GBS is present in the vagina and is thought to ascend into the uterus of some women where it can cause placental inflammation and preterm birth. Understanding the earliest events in the placenta that lead to preterm birth is elusive in humans, because the placenta cannot be studied until after birth. Here, we use a nonhuman primate model to show that an early GBS infection can damage the structural support of the fetal membranes, specifically the cytokeratin network in the epithelium of the amnion (one part of the membranes). Next, we obtained human placentas to show that this cytokeratin network was also damaged in human patients that had preterm premature rupture of the membranes, a major cause of preterm birth. Our work is important in understanding why fetal membranes may rupture prematurely, which may lead to early interventions to prevent membrane damage after placental infection and preterm birth.
Collapse
Affiliation(s)
- Jeroen P Vanderhoeven
- Department of Obstetrics & Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Craig J Bierle
- Center for Childhood Infections and Prematurity Research, Seattle Children's Research Institute, Seattle, Washington, United States of America
| | - Raj P Kapur
- Departments of Pathology, Seattle Children's and University of Washington, Seattle, Washington, United States of America
| | - Ryan M McAdams
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Richard P Beyer
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Federico M Farin
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Aasthaa Bansal
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Min Spencer
- Center on Human Development and Disability, University of Washington, Seattle, Washington, United States of America
| | - Mei Deng
- Center on Human Development and Disability, University of Washington, Seattle, Washington, United States of America
| | - Michael G Gravett
- Department of Obstetrics & Gynecology, University of Washington, Seattle, Washington, United States of America; Global Alliance to Prevent Prematurity & Stillbirth, Seattle, Washington, United States of America
| | - Craig E Rubens
- Center for Childhood Infections and Prematurity Research, Seattle Children's Research Institute, Seattle, Washington, United States of America; Department of Pediatrics, University of Washington, Seattle, Washington, United States of America; Global Alliance to Prevent Prematurity & Stillbirth, Seattle, Washington, United States of America
| | - Lakshmi Rajagopal
- Center for Childhood Infections and Prematurity Research, Seattle Children's Research Institute, Seattle, Washington, United States of America; Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
| | - Kristina M Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, Washington, United States of America; Center on Human Development and Disability, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
19
|
Li Y, Tian D. Correlation between apoptosis and TGF-β1 expression in the mucosal epithelium of rat small intestine in a cold stress state. Exp Ther Med 2013; 5:1456-1460. [PMID: 23737898 PMCID: PMC3671846 DOI: 10.3892/etm.2013.983] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 01/28/2013] [Indexed: 12/26/2022] Open
Abstract
The aim of this study was to investigate the correlation between the expression of transforming growth factor-β1 (TGF-β1) in the mucosal tissue of rat small intestine and the apoptosis of epithelial cells in the small intestine in a cold-restraint stress state. Immunohistochemistry was used to detect the expression of TGF-β1. Terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labeling (TUNEL) and DNA agarose gel electrophoresis were used to detect apoptosis. After 8 and 12 h of cold-restraint stress, the positive expression rate of TGF-β1 in the rat small intestine epithelial tissue was 59.09 and 54.16%, respectively. The apoptotic index (AI) of the rat small intestine epithelial cells was 25.69±8.09 and 19.65±6.61%, respectively. The positive expression rate of TGF-β1 in the epithelial tissue of the rat small intestine was positively correlated with the AI of the epithelial cells (r=0.980, P<0.05). The epithelial cells of the rat small intestine exhibited apoptosis under cold-restraint stress. TGF-β1 is one of the key factors that induces apoptosis of the epithelial cells of the rat small intestine.
Collapse
Affiliation(s)
- Yongjun Li
- Department of Gastroenterology, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832006
| | | |
Collapse
|
20
|
de Silva HC, Firth SM, Twigg SM, Baxter RC. Interaction between IGF binding protein-3 and TGFβ in the regulation of adipocyte differentiation. Endocrinology 2012; 153:4799-807. [PMID: 22910030 DOI: 10.1210/en.2011-1444] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The development of white adipose tissue involves both the hypertrophy of existing adipocytes and the proliferation and differentiation of preadipocytes. Adipogenic differentiation is inhibited by TGFβ signaling through Smad2/3, and IGF binding protein-3 (IGFBP-3) is also known to activate Smad2/3 signaling in some cell types. We previously reported that exogenous or overexpressed IGFBP-3 inhibits adipogenesis in 3T3-L1 cells, but the role of endogenous IGFBP-3 in this process, and its possible interaction with TGFβ, is not known. During 10-d adipogenic differentiation initiated by insulin, dexamethasone, and 3-isobutyl-1-methylxanthine, 3T3-L1 cells expressed increasing levels of IGFBP-3 and TGFβ1, secreting over 1000 pg/ml of both proteins. Exogenous recombinant human IGFBP-3 paralleled TGFβ1 in stimulating Smad2 phosphorylation in 3T3-L1 preadipocytes, but no additive effect was observed for the two agents. In contrast, knockdown of endogenous IGFBP-3 by small interfering RNA (siRNA) significantly impaired Smad2 activation by 0.25 ng/ml TGFβ1. Transient expression of human IGFBP-3 significantly inhibited the induction of adipogenic markers adiponectin and resistin, and the appearance of lipid droplets, but down-regulation of endogenous IGFBP-3 by siRNA had little effect on the expression of either marker during the 10-d differentiation, compared with nonsilencing control siRNA. However, down-regulation of endogenous IGFBP-3 using two different siRNA significantly reversed the inhibitory effect of TGFβ1 on both adiponectin and resistin induction. We conclude that IGFBP-3 activates inhibitory Smad signaling in 3T3-L1 cells and that endogenous IGFBP-3 modulates their adipogenic differentiation by regulating cell sensitivity towards the inhibitory effect of TGFβ.
Collapse
Affiliation(s)
- Hasanthi C de Silva
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales 2065, Australia
| | | | | | | |
Collapse
|
21
|
Zhao M, Sachs PC, Wang X, Dumur CI, Idowu MO, Robila V, Francis MP, Ware J, Beckman M, Rizki A, Holt SE, Elmore LW. Mesenchymal stem cells in mammary adipose tissue stimulate progression of breast cancer resembling the basal-type. Cancer Biol Ther 2012; 13:782-92. [PMID: 22669576 DOI: 10.4161/cbt.20561] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Data are accumulating to support a role for adipose-derived mesenchymal stem cells (MSCs) in breast cancer progression; however, to date most studies have relied on adipose MSCs from non-breast sources. There is a particular need to investigate the role of adipose MSCs in the pathogenesis of basal-like breast cancer, which develops at a disproportionate rate in pre-menopausal African-American women with a gain in adiposity. The aim of this study was to better understand how breast adipose MSCs (bMSCs) contribute to the progression of basal-like breast cancers by relying on isogenic HMT-3255 S3 (pre-invasive) and T4-2 (invasive) human cells that upon transplantation into nude mice resemble this tumor subtype. In vitro results suggested that bMSCs may contribute to breast cancer progression in multiple ways. bMSCs readily penetrate extracellular matrix components in part through their expression of matrix metalloproteinases 1 and 3, promote the invasion of T4-2 cells and efficiently chemoattract endothelial cells via a bFGF-independent, VEGF-A-dependent manner. As mixed xenografts, bMSCs stimulated the growth, invasion and desmoplasia of T4-2 tumors, yet these resident stem cells showed no observable effect on the progression of pre-invasive S3 cells. While bMSCs form vessel-like structures within Matrigel both in vitro and in vivo and chemoattract endothelial cells, there appeared to be no difference between T4-2/bMSC mixed xenografts and T4-2 xenografts with regard to intra- or peri-tumoral vascularity. Collectively, our data suggest that bMSCs may contribute to the progression of basal-like breast cancers by stimulating growth and invasion but not vasculogenesis or angiogenesis.
Collapse
Affiliation(s)
- Min Zhao
- Department of Pathology, Virginia Commonwealth University, Richmond, Virginia, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Petit JY, Botteri E, Lohsiriwat V, Rietjens M, De Lorenzi F, Garusi C, Rossetto F, Martella S, Manconi A, Bertolini F, Curigliano G, Veronesi P, Santillo B, Rotmensz N. Locoregional recurrence risk after lipofilling in breast cancer patients. Ann Oncol 2012; 23:582-588. [PMID: 21610155 DOI: 10.1093/annonc/mdr158] [Citation(s) in RCA: 161] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Lipofilling has been indicated for postmastectomy and postlumpectomy breast reconstruction. The clinical literatures underline its technical efficacy but experimental studies raise important questions about the potential detrimental effect of adipocytes on the stimulation of cancer growth and reappearance. DESIGN We collected 321 consecutive patients operated for a primary breast cancer between 1997 and 2008 who subsequently underwent lipofilling for reconstructive purpose. For each patient, we selected two matched patients with similar characteristics who did not undergo a lipofilling. RESULTS Eighty-nine percent of the tumors were invasive. Median follow-up was 56 months from the primary surgery and 26 months from the lipofilling. Eight and 19 patients had a local event in the lipofilling and control group, respectively, leading to comparable cumulative incidence curves [P = 0.792; Hazard Ratio(Lipo vs No lipo) = 1.11 (95% confidence interval 0.47-2.64)]. These results were confirmed when patients undergoing quadrantectomy and mastectomy were analyzed separately and when the analysis was limited to invasive tumors. Based on 37 cases, the lipofilling group resulted at higher risk of local events when the analysis was limited to intraepithelial neoplasia. CONCLUSIONS Lipofilling seems to be a safe procedure in breast cancer patients. Longer follow-up and further experiences from oncological series are urgently required to confirm these findings.
Collapse
Affiliation(s)
| | - E Botteri
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - V Lohsiriwat
- Division of Plastic Surgery; Department of Surgery, Mahidol University, Siriraj Hospital, Bangkok, Thailand
| | | | | | | | | | | | | | | | - G Curigliano
- Division of Medical Oncology, Department of Medicine
| | - P Veronesi
- Division of Breast Surgery, European Institute of Oncology, Milan; University of Milan, Milan, Italy
| | - B Santillo
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - N Rotmensz
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| |
Collapse
|
23
|
The oncologic outcome and immediate surgical complications of lipofilling in breast cancer patients: a multicenter study--Milan-Paris-Lyon experience of 646 lipofilling procedures. Plast Reconstr Surg 2011; 128:341-346. [PMID: 21502905 DOI: 10.1097/prs.0b013e31821e713c] [Citation(s) in RCA: 188] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Lipofilling is now performed to improve the breast contour, after both breast-conserving surgery and breast reconstruction. However, injection of fat into a previous tumor site may create a new environment for cancer and adjacent cells. There is also no international agreement regarding lipofilling after breast cancer treatment. METHODS The authors included three institutions specializing in both breast cancer treatment and breast reconstruction (European Institute of Oncology, Milan, Italy; Paris Breast Center, Paris, France; and Leon Berard Centre, Lyon, France) for a multicenter study. A collective chart review of all lipofilling procedures after breast cancer treatment was performed. RESULTS From 2000 to 2010, the authors reviewed 646 lipofilling procedures from 513 patients. There were 370 mastectomy patients and 143 breast-conserving surgery patients. There were 405 patients (78.9 percent) with invasive carcinoma and 108 (21.1 percent) with carcinoma in situ. The average interval between oncologic surgical interventions and lipofilling was 39.7 months. Average follow-up after lipofilling was 19.2 months. The authors observed a complication rate of 2.8 percent (liponecrosis, 2.0 percent). Twelve radiologic images appeared after lipofilling in 119 breast-conserving surgery cases (10.1 percent). The overall oncologic event rate was 5.6 percent (3.6 percent per year). The locoregional event rate was 2.4 percent (1.5 percent per year). CONCLUSIONS Lipofilling after breast cancer treatment leads to a low complication rate and does not affect radiologic follow-up after breast-conserving surgery. A prospective clinical registry including high-volume multicenter data with a long follow-up is warranted to demonstrate the oncologic safety. Until then, lipofilling should be performed in experienced hands, and a cautious oncologic follow-up protocol is advised. CLINICAL QUESTION/LEVEL OF EVIDENCE Therapeutic, IV [corrected].
Collapse
|
24
|
Autologous fat transplantation in patients with breast cancer: “silencing” or “fueling” cancer recurrence? Breast 2011; 20:351-7. [DOI: 10.1016/j.breast.2011.01.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 09/15/2010] [Accepted: 01/06/2011] [Indexed: 01/22/2023] Open
|
25
|
Rietjens M, De Lorenzi F, Rossetto F, Brenelli F, Manconi A, Martella S, Intra M, Venturino M, Lohsiriwat V, Ahmed Y, Petit J. Safety of fat grafting in secondary breast reconstruction after cancer. J Plast Reconstr Aesthet Surg 2011; 64:477-83. [DOI: 10.1016/j.bjps.2010.06.024] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Revised: 06/22/2010] [Accepted: 06/28/2010] [Indexed: 01/15/2023]
|
26
|
Gnedeva KY, Chermnykh ES, Vorotelyak EA, Vasil’ev AV, Terskikh VV. Effect of growth factors on morphogenesis of human keratinocytes in vitro. BIOL BULL+ 2009. [DOI: 10.1134/s106235900903011x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
27
|
Wang H, Li M, Lin PH, Yao Q, Chen C. Fluid shear stress regulates the expression of TGF-beta1 and its signaling molecules in mouse embryo mesenchymal progenitor cells. J Surg Res 2008; 150:266-70. [PMID: 18547588 PMCID: PMC2927960 DOI: 10.1016/j.jss.2007.12.801] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 12/19/2007] [Accepted: 12/19/2007] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recently we reported that fluid shear stress promotes endothelial cell differentiation from a mouse embryo mesenchymal progenitor cell line C3H10T1/2. However, it is not clear whether the transforming growth factor-beta 1 (TGF-beta1) system is associated with shear-induced endothelial differentiation. The purpose of this study was to determine the effect of shear stress on the expression of TGF-beta1 and its signaling molecules in C3H10T1/2 cells. METHODS Murine C3H10T1/2 cells were incubated on collagen Type 1-coated dishes, and subjected to a steady fluid shear stress of 15 dyn/cm(2) for 6, 12, and 24 h. The mRNA levels for TGF-beta1, TGF-beta receptors (TGF-beta R), and Smad molecules were determined with real-time PCR analysis and normalized to glyceraldehyde-3-phosphate dehydrogenase mRNA levels. RESULTS TGF-beta1 mRNA expression was down-regulated by 60% and 66% in shear stress-treated cells at 12 and 24 h, respectively, compared with static control group (P < 0.01). In addition, shear stress significantly decreased TGF-beta R1 mRNA levels by 30% and 50% in shear stress-treated cells at 12 and 24 h, respectively (P < 0.01). For TGF-beta R2, shear stress at 6, 12, and 24 h significantly reduced its expression by 93%, 95% and 97%, respectively, compared with static controls (P < 0.01). Furthermore, shear stress significant decreased mRNA levels of positive signaling molecules Smad2, Smad3, and Smad4 in a time-dependent manner (P < 0.01). However, shear stress significantly increased negative signaling molecule Smad7 mRNA levels by 100% at 24 h treatment compared with static control group (P < 0.01). CONCLUSIONS Fluid shear stress significantly suppresses TGF-beta1 functions through down-regulation of TGF-beta1, TGF-beta R, positive signaling molecules Smad2, Smad3, Smad4, and up-regulation of negative signaling molecule Smad7 in a mouse embryo mesenchymal progenitor cell line C3H10T1/2. This study suggests that the negative regulation of the TGF-beta1 system may be involved in shear-induced endothelial cell differentiation in C3H10T1/2.
Collapse
Affiliation(s)
- Hao Wang
- Molecular Surgeon Research Center, Division of Vascular Surgery and Endovascular Therapy, Michael E. DeBakey Department of Surgery, Michael E. DeBakey VA Medical Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
28
|
Horie T, Ono K, Kinoshita M, Nishi H, Nagao K, Kawamura T, Abe Y, Wada H, Shimatsu A, Kita T, Hasegawa K. TG-interacting factor is required for the differentiation of preadipocytes. J Lipid Res 2008; 49:1224-34. [PMID: 18310014 DOI: 10.1194/jlr.m700578-jlr200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The accumulation of visceral adipose tissue is closely associated with insulin resistance and metabolic syndrome. Therefore, it is important to identify genes that are required for adipocyte differentiation. To identify genes that are required for the differentiation of 3T3-L1 preadipocytes into mature adipocytes, we used retrovirus insertion-mediated random mutagenesis to generate 3T3-L1 cell lines that lose their ability to differentiate into mature adipocytes. One of the genes identified was TG-interacting factor (TGIF), a DNA binding homeodomain protein that has been demonstrated to suppress Smad-mediated activation of transforming growth factor beta (TGF-beta)-regulated transcription. In the TGIF-disrupted clone of 3T3-L1 preadipocytes, the rate of differentiation into mature adipocytes was clearly reduced compared with that in the wild-type clone. Suppression of TGIF by lentivirus-mediated RNAi also inhibited the differentiation of 3T3-L1 cells. Insulin specifically increased the abundance of TGIF protein, primarily by enhancing its stability. In addition, insulin caused the rapid accumulation of TGIF in the nuclei. Forced expression of exogenous TGIF repressed both endogenous and overexpressed Smad2/3-mediated promoter activity in 3T3-L1. These findings suggest that insulin specifically antagonizes TGF-beta signaling in preadipocytes by stabilizing the putative Smad transcriptional corepressor TGIF and regulates adipocyte differentiation.
Collapse
Affiliation(s)
- Takahiro Horie
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Improved knowledge of all aspects of adipose biology will be required to counter the burgeoning epidemic of obesity. Interest in adipogenesis has increased markedly over the past few years with emphasis on the intersection between extracellular signals and the transcriptional cascade that regulates adipocyte differentiation. Many different events contribute to the commitment of a mesenchymal stem cell to the adipocyte lineage including the coordination of a complex network of transcription factors, cofactors and signalling intermediates from numerous pathways.
Collapse
Affiliation(s)
- Evan D Rosen
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.
| | | |
Collapse
|
30
|
Fain JN, Tichansky DS, Madan AK. Transforming growth factor beta1 release by human adipose tissue is enhanced in obesity. Metabolism 2005; 54:1546-51. [PMID: 16253647 DOI: 10.1016/j.metabol.2005.05.024] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2005] [Accepted: 05/23/2005] [Indexed: 01/04/2023]
Abstract
The present studies examined the effect of obesity in humans on the release of transforming growth factor beta1 (TGF-beta1) by human adipose tissue. The regulation of TGF-beta1 release by adipose tissue as well as the question of whether its release is due to the adipocytes or the nonfat cells in adipose tissue was also examined. There was a statistically significant (r=0.50) correlation between the body mass index of the fat donors and the subsequent release of TGF-beta1 release by subcutaneous adipose tissue. There was also a positive correlation between total TGF-beta1 release by adipose tissue explants and body fat content (r=0.69). The question of whether tumor necrosis factor alpha (TNF-alpha) and/or interleukin 1 beta (IL-1 beta) regulate the release of TGF-beta1 was investigated by incubation of adipose tissue explants with a soluble human TNF-alpha receptor (etanercept) and a neutralizing antihuman IL-1 beta antibody. The release of TGF-beta1 over 48 hours by adipose tissue explants was significantly enhanced in the presence of both the inhibitor of TNF-alpha and of IL-1 beta. It is of interest, in view of the elevated circulating insulin in blood of morbidly obese women, that the release of TGF-beta1 by adipose tissue was enhanced in the presence of insulin. The question of whether the release of TGF-beta1 by human adipose tissue explants was primarily due to adipocytes, as is the case for leptin, or the nonfat cells present in human adipose tissue, as is the case for IL-8 and prostaglandin E(2), was examined. The release of TGF-beta1 was primarily by the nonfat cells of human adipose tissue because release by adipocytes was less than 10% of that by the nonfat cells of adipose tissue.
Collapse
Affiliation(s)
- John N Fain
- Department of Molecular Sciences, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | | | | |
Collapse
|
31
|
Zhao H, Langerød A, Ji Y, Nowels KW, Nesland JM, Tibshirani R, Bukholm IK, Kåresen R, Botstein D, Børresen-Dale AL, Jeffrey SS. Different gene expression patterns in invasive lobular and ductal carcinomas of the breast. Mol Biol Cell 2004; 15:2523-36. [PMID: 15034139 PMCID: PMC420079 DOI: 10.1091/mbc.e03-11-0786] [Citation(s) in RCA: 321] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Invasive ductal carcinoma (IDC) and invasive lobular carcinoma (ILC) are the two major histological types of breast cancer worldwide. Whereas IDC incidence has remained stable, ILC is the most rapidly increasing breast cancer phenotype in the United States and Western Europe. It is not clear whether IDC and ILC represent molecularly distinct entities and what genes might be involved in the development of these two phenotypes. We conducted comprehensive gene expression profiling studies to address these questions. Total RNA from 21 ILCs, 38 IDCs, two lymph node metastases, and three normal tissues were amplified and hybridized to approximately 42,000 clone cDNA microarrays. Data were analyzed using hierarchical clustering algorithms and statistical analyses that identify differentially expressed genes (significance analysis of microarrays) and minimal subsets of genes (prediction analysis for microarrays) that succinctly distinguish ILCs and IDCs. Eleven of 21 (52%) of the ILCs ("typical" ILCs) clustered together and displayed different gene expression profiles from IDCs, whereas the other ILCs ("ductal-like" ILCs) were distributed between different IDC subtypes. Many of the differentially expressed genes between ILCs and IDCs code for proteins involved in cell adhesion/motility, lipid/fatty acid transport and metabolism, immune/defense response, and electron transport. Many genes that distinguish typical and ductal-like ILCs are involved in regulation of cell growth and immune response. Our data strongly suggest that over half the ILCs differ from IDCs not only in histological and clinical features but also in global transcription programs. The remaining ILCs closely resemble IDCs in their transcription patterns. Further studies are needed to explore the differences between ILC molecular subtypes and to determine whether they require different therapeutic strategies.
Collapse
Affiliation(s)
- Hongjuan Zhao
- Department of Surgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Iyengar P, Combs TP, Shah SJ, Gouon-Evans V, Pollard JW, Albanese C, Flanagan L, Tenniswood MP, Guha C, Lisanti MP, Pestell RG, Scherer PE. Adipocyte-secreted factors synergistically promote mammary tumorigenesis through induction of anti-apoptotic transcriptional programs and proto-oncogene stabilization. Oncogene 2003; 22:6408-23. [PMID: 14508521 DOI: 10.1038/sj.onc.1206737] [Citation(s) in RCA: 267] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mammary epithelial cells are embedded in a unique extracellular environment to which adipocytes and other stromal cells contribute. Mammary epithelial cells are critically dependent on this milieu for survival. However, it remains unknown which adipocyte-secreted factors are required for the survival of the mammary epithelia and what role these adipokines play in the process of ductal carcinoma tumorigenesis. Here, we take a systematic molecular approach to investigate the multiple ways adipocytes and adipokines can uniquely influence the characteristics and phenotypic behavior of malignant breast ductal epithelial cells. Microarray analysis and luciferase reporter assays indicate that adipokines specifically induce several transcriptional programs involved in promoting tumorigenesis, including increased cell proliferation (IGF2, FOS, JUN, cyclin D1), invasive potential (MMP1, ATF3), survival (A20, NFkappaB), and angiogenesis. One of the key changes in the transformed ductal epithelial cells associated with the cell cycle involves the induction of NFkappaB (five-fold) and cyclin D1 (three-fold). We show that by regulating the transcription of these molecules, the synergistic activity of adipocyte-derived factors can potentiate MCF-7 cell proliferation. Furthermore, compared to other stromal cell-secreted factors, the full complement of adipokines shows an unparalleled ability to promote increased cell motility, migration, and the capacity for angiogenesis. Adipocyte-secreted factors can affect tumorigenesis by increasing the stabilization of pro-oncogenic factors such as beta-catenin and CDK6 as a result of a reduction in the gene expression of their inhibitors (i.e. p18). An in vivo coinjection system using 3T3-L1 adipocytes and SUM159PT cells effectively recapitulates the host-tumor interactions in primary tumors. Type VI collagen, a soluble extracellular matrix protein abundantly expressed in adipocytes, is further upregulated in adipocytes during tumorigenesis. It promotes GSK3beta phosphorylation, beta-catenin stabilization, and increased beta-catenin activity in breast cancer cells and may critically contribute towards tumorigenesis when not counterbalanced by other factors.
Collapse
Affiliation(s)
- Puneeth Iyengar
- Department of Cell Biology, Albert Einstein Cancer Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chen HH, Zhao S, Song JG. TGF-beta1 suppresses apoptosis via differential regulation of MAP kinases and ceramide production. Cell Death Differ 2003; 10:516-27. [PMID: 12728250 DOI: 10.1038/sj.cdd.4401171] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Serum deprivation induces apoptosis in NIH3T3 cells, which is associated with increased intracellular ceramide generation and with the activation of p38 mitogen-activated protein (MAP) kinase. Treatment of cells with transforming growth factor-beta1 (TGF-beta1) activated the extracellular signal regulated kinases 1 and 2 (ERK1/ERK2), inhibited the serum deprivation-induced p38 activation and the increase in intracellular ceramide formation, leading to the stimulation of cell proliferation and the suppression of apoptosis. Inhibition of p38 MAP kinase by SB203580 significantly reduced the serum-deprivation-induced apoptosis. Overexpression of p38 increased the cell apoptosis and reduced the antiapoptotic effect of TGF-beta1. Inhibition of ERK1/ERK2 by PD98059 completely inhibited the TGF-beta1-stimulated proliferation and partially inhibited the antiapoptotic effects of TGF-beta1. Neither SB203580 nor PD98059 has obvious effect on TGF-beta1-mediated inhibition of the increased ceramide generation. Serum-deprivation-induced apoptosis in NIH3T3 cells can also be blocked by broad-spectrum caspase inhibitor. TGF-beta1 treatment has an inhibitory effect on caspase activities. Our results indicate that ceramide, p38, and ERK1/ERK2 play critical but differential roles in cell proliferation and stress-induced apoptosis. TGF-beta1 suppresses the serum-deprivation-induced apoptosis via its distinct effects on complex signaling events involving the activation of ERK1/ERK2 and the inhibition of p38 activation and increased ceramide generation.
Collapse
Affiliation(s)
- H-H Chen
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | | | | |
Collapse
|
34
|
Choy L, Derynck R. Transforming growth factor-beta inhibits adipocyte differentiation by Smad3 interacting with CCAAT/enhancer-binding protein (C/EBP) and repressing C/EBP transactivation function. J Biol Chem 2003; 278:9609-19. [PMID: 12524424 DOI: 10.1074/jbc.m212259200] [Citation(s) in RCA: 274] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor (TGF)-beta is a potent inhibitor of adipocyte differentiation. To identify which adipocyte transcription factors might be targeted by TGF-beta, we overexpressed key adipogenic transcription factors, C/EBPbeta, C/EBPdelta, or peroxisome proliferator-activated receptor (PPAR) gamma in NIH3T3 cells and tested the ability of TGF-beta to block adipogenesis. We show that TGF-beta inhibits adipocyte differentiation driven by either C/EBPbeta or C/EBPdelta without affecting C/EBP protein expression levels, suggesting that these C/EBPs are a direct target of TGF-beta action. Because TGF-beta inhibits adipogenesis by signaling through Smad3, we examined physical and functional interactions of Smad3 and Smad4 with C/EBPbeta, C/EBPdelta, and PPARgamma2. C/EBPbeta and C/EBPdelta were found to physically interact with Smad3 and Smad4, and Smad3 cooperated with Smad4 and TGF-beta signaling to repress the transcriptional activity of C/EBPs. Thus, repression of the activity of C/EBPs by Smad3/4 at C/EBP binding sites inhibited transcription from the PPARgamma2 and leptin promoters. In contrast, PPARgamma interacted only very weakly with Smad3 and its transcriptional activity was not repressed by Smad3/4 or in response to TGF-beta. Smad3/4 did not reduce the ability of C/EBP to bind to its cognate DNA sequence, but repressed transcription by inhibiting the transactivation function of C/EBP.
Collapse
Affiliation(s)
- Lisa Choy
- Department of Growth and Development, Programs in Cell Biology and Developmental Biology, University of California, San Francisco, California 94143-0640, USA
| | | |
Collapse
|
35
|
Imagawa W, Pedchenko VK, Helber J, Zhang H. Hormone/growth factor interactions mediating epithelial/stromal communication in mammary gland development and carcinogenesis. J Steroid Biochem Mol Biol 2002; 80:213-30. [PMID: 11897505 DOI: 10.1016/s0960-0760(01)00188-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Epithelial/mesenchymal interactions begin during embryonic development of the mammary gland and continue throughout mammary gland development into adult life. Stromal and epithelial growth factors that may mediate interactions between these compartments of the mammary gland are reviewed. Since mammogenic hormones are the primary regulators of mammary gland development, special consideration is given to hormonal regulation of growth factors in order to explore the integration of hormones and growth factors in the regulation of mammary gland growth and neoplasia. Examination of hormonal regulation of the fibroblast growth factor (FGF)-7/FGFR2-IIIb receptor system in the mammary gland reveals that mammogenic hormones differentially regulate the synthesis of stromal growth factors and their epithelial receptors. These effects serve to optimize the action of estrogen and progesterone on mammary gland development and illustrate that the ratio of these two hormones is critical in regulating this growth factor axis. The role of stromal/epithelial mitogenic microenvironments in modulating the genotype and phenotype of preneoplastic and neoplastic lesions by chemical carcinogens is discussed. Finally, changes in growth factor expression during mammary tumor progression are described to illustrate the relative roles that stromally-derived and epithelial-derived growth factors may play during progression to hormone independent tumor growth.
Collapse
Affiliation(s)
- Walter Imagawa
- Department of Molecular and Integrative Physiology, Kansas Cancer Institute, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160-7417, USA.
| | | | | | | |
Collapse
|
36
|
Blokzijl A, ten Dijke P, Ibáñez CF. Physical and functional interaction between GATA-3 and Smad3 allows TGF-beta regulation of GATA target genes. Curr Biol 2002; 12:35-45. [PMID: 11790301 DOI: 10.1016/s0960-9822(01)00623-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Members of the GATA family of zinc finger transcription factors are genetically controlled "master" regulators of development in the hematopoietic and nervous systems. Whether GATA factors also serve to integrate epigenetic signals on target promoters is, however, unknown. The transforming growth factor-beta (TGF-beta) superfamily is a large group of phylogenetically conserved secreted factors controlling cell proliferation, differentiation, migration, and survival in multiple tissues. RESULTS GATA-3, a key regulator of T helper cell development, was found to directly interact with Smad3, an intracellular signal transducer of TGF-beta. Complex formation required a central region in GATA-3 and the N-terminal domain of Smad3. GATA-3 mediated recruitment of Smad3 to GATA binding sites independently of Smad3 binding to DNA, and the two factors cooperated synergistically to regulate transcription from the IL-5 promoter in a TGF-beta-dependent manner. Treatment of T helper cells with TGF-beta promoted the formation of an endogenous Smad3/GATA-3 nuclear complex and stimulated production of the Th2 cytokine IL-10 in a Smad3- and GATA-3-dependent manner. CONCLUSIONS Although Smad proteins are known to interact with a number of general transcription factors, these are insufficient to explain the tissue-specific biology of TGF-beta proteins. Through its interaction with Smad3, GATA-3 is able to integrate a genetic program of cell differentiation with an extracellular signal, providing a molecular framework for the effects of TGF-beta on the development and function of specific subsets of immune cells and possibly other cell types.
Collapse
Affiliation(s)
- Andries Blokzijl
- Division of Molecular Neurobiology, Department of Neuroscience, Karolinska Institute, 171 77, Stockholm, Sweden
| | | | | |
Collapse
|
37
|
Seo HS, Larsimont D, Ma Y, Laios I, Leclercq G. Regulation of estrogen receptor levels by ligand-induced release of compound(s) in MCF-7 cells. Mol Cell Endocrinol 2000; 164:19-29. [PMID: 11026554 DOI: 10.1016/s0303-7207(00)00244-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In MCF-7 cells, estradiol (E2) and pure antiestrogens (AEs) decrease estrogen receptor alpha (ER) levels, while AEs with partial estrogenic activity lead to ER alpha accumulation. Using immunocytochemistry, we found that cells pre-exposed to one of such ligands, when plated with untreated cells, led to similar ER changes in the latter. Conditioned media (CMs) prepared from stimulated cells displayed identical regulatory effects even after strong dilution; they also modulated ERE-dependent transcriptional activity. Evaluation of residual ligand concentrations in CMs rejected the possibility of a major interference of the former. Cycloheximide, which inhibits E2-induced down-regulation, failed to block the influence of CM(E2) in agreement with this view. DCC-treatment of CMs abrogated their effects, suggesting the release of hydrophobic compound(s) which regulate ER and/or amplify the effect of extremely low amounts of residual ligands. Such a release appears independent of ER since CMs from MDA-MB-231 cells (ER-negative) were effective as their autologous media on MCF-7 cells.
Collapse
Affiliation(s)
- H S Seo
- Laboratoire J.C. Heuson de Canérologie Mammaire, Service de Médecine Interne, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | |
Collapse
|
38
|
Choy L, Skillington J, Derynck R. Roles of autocrine TGF-beta receptor and Smad signaling in adipocyte differentiation. J Cell Biol 2000; 149:667-82. [PMID: 10791980 PMCID: PMC2174852 DOI: 10.1083/jcb.149.3.667] [Citation(s) in RCA: 253] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
TGF-beta inhibits adipocyte differentiation, yet is expressed by adipocytes. The function of TGF-beta in adipogenesis, and its mechanism of action, is unknown. To address the role of TGF-beta signaling in adipocyte differentiation, we characterized the expression of the TGF-beta receptors, and the Smads which transmit or inhibit TGF-beta signals, during adipogenesis in 3T3-F442A cells. We found that the cell-surface availability of TGF-beta receptors strongly decreased as adipogenesis proceeds. Whereas mRNA levels for Smads 2, 3, and 4 were unchanged during differentiation, mRNA levels for Smads 6 and 7, which are known to inhibit TGF-beta responses, decreased severely. Dominant negative interference with TGF-beta receptor signaling, by stably expressing a truncated type II TGF-beta receptor, enhanced differentiation and decreased growth. Stable overexpression of Smad2 or Smad3 inhibited differentiation and dominant negative inhibition of Smad3 function, but not Smad2 function, enhanced adipogenesis. Increased Smad6 and Smad7 levels blocked differentiation and enhanced TGF-beta-induced responses. The inhibitory effect of Smad7 on adipocyte differentiation and its cooperation with TGF-beta was associated with the C-domain of Smad7. Our results indicate that endogenous TGF-beta signaling regulates the rate of adipogenesis, and that Smad2 and Smad3 have distinct functions in this endogenous control of differentiation. Smad6 and Smad7 act as negative regulators of adipogenesis and, even though known to inhibit TGF-beta responses, enhance the effects of TGF-beta on these cells.
Collapse
Affiliation(s)
- Lisa Choy
- Department of Growth and Development, Programs in Cell Biology and Developmental Biology, University of California at San Francisco, San Francisco, California 94143-0640
- Department of Anatomy, Programs in Cell Biology and Developmental Biology, University of California at San Francisco, San Francisco, California 94143-0640
| | - Jeremy Skillington
- Department of Growth and Development, Programs in Cell Biology and Developmental Biology, University of California at San Francisco, San Francisco, California 94143-0640
- Department of Anatomy, Programs in Cell Biology and Developmental Biology, University of California at San Francisco, San Francisco, California 94143-0640
| | - Rik Derynck
- Department of Growth and Development, Programs in Cell Biology and Developmental Biology, University of California at San Francisco, San Francisco, California 94143-0640
- Department of Anatomy, Programs in Cell Biology and Developmental Biology, University of California at San Francisco, San Francisco, California 94143-0640
| |
Collapse
|
39
|
Barcellos-Hoff MH, Ewan KB. Transforming growth factor-beta and breast cancer: Mammary gland development. Breast Cancer Res 2000; 2:92-9. [PMID: 11250698 PMCID: PMC139430 DOI: 10.1186/bcr40] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/1999] [Revised: 01/31/2000] [Accepted: 02/01/2000] [Indexed: 11/13/2022] Open
Abstract
Transforming growth factor (TGF)-beta1 is a pluripotent cytokine that profoundly inhibits epithelial proliferation, induces apoptosis, and influences morphogenesis by mediating extracellular matrix deposition and remodeling. The physiologic roles of the action of TGF-beta in mammary gland, indeed in most tissues, are poorly understood. In order to understand the actions of TGF-beta, we need to take into account the complexity of its effects on different cell types and the influence of context on cellular responses. This task is further compounded by multiple mechanisms for regulating TGF-beta transcription, translation, and activity. One of the most significant factors that obscures the action of TGF-beta is that it is secreted as a stable latent complex, which consists of the 24-kDa cytokine and the 80-kDa dimer of its prepro region, called latency-associated peptide. Latency imposes a critical restraint on TGF-beta activity that is often overlooked. The extracellular process known as activation, in which TGF-beta is released from the latent complex, is emphasized in the present discussion of the role of TGF-beta in mammary gland development. Definition of the spatial and temporal patterns of latent TGF-beta activation in situ is essential for understanding the specific roles that TGF-beta plays during mammary gland development, proliferation, and morphogenesis.
Collapse
Affiliation(s)
- M H Barcellos-Hoff
- Life Sciences Division, Lawrence Berkeley National Laboratory, University of California, Berkeley, CA 94720, USA.
| | | |
Collapse
|