1
|
Luca T, Pezzino S, Puleo S, Castorina S. Lesson on obesity and anatomy of adipose tissue: new models of study in the era of clinical and translational research. J Transl Med 2024; 22:764. [PMID: 39143643 PMCID: PMC11323604 DOI: 10.1186/s12967-024-05547-3] [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: 03/10/2024] [Accepted: 07/28/2024] [Indexed: 08/16/2024] Open
Abstract
Obesity is a serious global illness that is frequently associated with metabolic syndrome. Adipocytes are the typical cells of adipose organ, which is composed of at least two different tissues, white and brown adipose tissue. They functionally cooperate, interconverting each other under physiological conditions, but differ in their anatomy, physiology, and endocrine functions. Different cellular models have been proposed to study adipose tissue in vitro. They are also useful for elucidating the mechanisms that are responsible for a pathological condition, such as obesity, and for testing therapeutic strategies. Each cell model has its own characteristics, culture conditions, advantages and disadvantages. The choice of one model rather than another depends on the specific study the researcher is conducting. In recent decades, three-dimensional cultures, such as adipose spheroids, have become very attractive because they more closely resemble the phenotype of freshly isolated cells. The use of such models has developed in parallel with the evolution of translational research, an interdisciplinary branch of the biomedical field, which aims to learn a scientific translational approach to improve human health and longevity. The focus of the present review is on the growing body of data linking the use of new cell models and the spread of translational research. Also, we discuss the possibility, for the future, to employ new three-dimensional adipose tissue cell models to promote the transition from benchside to bedsite and vice versa, allowing translational research to become routine, with the final goal of obtaining clinical benefits in the prevention and treatment of obesity and related disorders.
Collapse
Affiliation(s)
- Tonia Luca
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy.
| | | | - Stefano Puleo
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| | - Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| |
Collapse
|
2
|
Laber S, Strobel S, Mercader JM, Dashti H, dos Santos FR, Kubitz P, Jackson M, Ainbinder A, Honecker J, Agrawal S, Garborcauskas G, Stirling DR, Leong A, Figueroa K, Sinnott-Armstrong N, Kost-Alimova M, Deodato G, Harney A, Way GP, Saadat A, Harken S, Reibe-Pal S, Ebert H, Zhang Y, Calabuig-Navarro V, McGonagle E, Stefek A, Dupuis J, Cimini BA, Hauner H, Udler MS, Carpenter AE, Florez JC, Lindgren C, Jacobs SB, Claussnitzer M. Discovering cellular programs of intrinsic and extrinsic drivers of metabolic traits using LipocyteProfiler. CELL GENOMICS 2023; 3:100346. [PMID: 37492099 PMCID: PMC10363917 DOI: 10.1016/j.xgen.2023.100346] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 08/22/2022] [Accepted: 05/26/2023] [Indexed: 07/27/2023]
Abstract
A primary obstacle in translating genetic associations with disease into therapeutic strategies is elucidating the cellular programs affected by genetic risk variants and effector genes. Here, we introduce LipocyteProfiler, a cardiometabolic-disease-oriented high-content image-based profiling tool that enables evaluation of thousands of morphological and cellular profiles that can be systematically linked to genes and genetic variants relevant to cardiometabolic disease. We show that LipocyteProfiler allows surveillance of diverse cellular programs by generating rich context- and process-specific cellular profiles across hepatocyte and adipocyte cell-state transitions. We use LipocyteProfiler to identify known and novel cellular mechanisms altered by polygenic risk of metabolic disease, including insulin resistance, fat distribution, and the polygenic contribution to lipodystrophy. LipocyteProfiler paves the way for large-scale forward and reverse deep phenotypic profiling in lipocytes and provides a framework for the unbiased identification of causal relationships between genetic variants and cellular programs relevant to human disease.
Collapse
Affiliation(s)
- Samantha Laber
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Sophie Strobel
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
| | - Josep M. Mercader
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Hesam Dashti
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Felipe R.C. dos Santos
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Phil Kubitz
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Else Kröner-Fresenius-Centre for Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Maya Jackson
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alina Ainbinder
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Julius Honecker
- Else Kröner-Fresenius-Centre for Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
| | - Saaket Agrawal
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Garrett Garborcauskas
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David R. Stirling
- Imaging Platform, Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Aaron Leong
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Katherine Figueroa
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nasa Sinnott-Armstrong
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Genetics, Stanford University, San Francisco, CA, USA
| | - Maria Kost-Alimova
- Imaging Platform, Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Giacomo Deodato
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alycen Harney
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gregory P. Way
- Imaging Platform, Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alham Saadat
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sierra Harken
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Saskia Reibe-Pal
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK
| | - Hannah Ebert
- Institute of Nutritional Science, University Hohenheim, 70599 Stuttgart, Germany
| | - Yixin Zhang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Virtu Calabuig-Navarro
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Institute of Nutritional Science, University Hohenheim, 70599 Stuttgart, Germany
| | - Elizabeth McGonagle
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Adam Stefek
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC H3A 1G1, Canada
| | - Beth A. Cimini
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hans Hauner
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
- Else Kröner-Fresenius-Centre for Nutritional Medicine, School of Life Sciences, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Miriam S. Udler
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Anne E. Carpenter
- Imaging Platform, Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jose C. Florez
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Cecilia Lindgren
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Suzanne B.R. Jacobs
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Melina Claussnitzer
- Programs in Metabolism and Medical and Population Genetics, Type 2 Diabetes Systems Genomics Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
3
|
Neto IVDS, Pinto AP, Muñoz VR, de Cássia Marqueti R, Pauli JR, Ropelle ER, Silva ASRD. Pleiotropic and multi-systemic actions of physical exercise on PGC-1α signaling during the aging process. Ageing Res Rev 2023; 87:101935. [PMID: 37062444 DOI: 10.1016/j.arr.2023.101935] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/10/2023] [Accepted: 04/13/2023] [Indexed: 04/18/2023]
Abstract
Physical training is a potent therapeutic approach for improving mitochondrial health through peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) signaling pathways. However, comprehensive information regarding the physical training impact on PGC-1α in the different physiological systems with advancing age is not fully understood. This review sheds light on the frontier-of-knowledge data regarding the chronic effects of exercise on the PGC-1α signaling pathways in rodents and humans. We address the molecular mechanisms involved in the different tissues, clarifying the precise biological action of PGC-1α, restricted to the aged cell type. Distinct exercise protocols (short and long-term) and modalities (aerobic and resistance exercise) increase the transcriptional and translational PGC-1α levels in adipose tissue, brain, heart, liver, and skeletal muscle in animal models, suggesting that this versatile molecule induces pleiotropic responses. However, PGC-1α function in some human tissues (adipose tissue, heart, and brain) remains challenging for further investigations. PGC-1α is not a simple transcriptional coactivator but supports a biochemical environment of mitochondrial dynamics, controlling physiological processes (primary metabolism, tissue remodeling, autophagy, inflammation, and redox balance). Acting as an adaptive mechanism, the long-term effects of PGC-1α following exercise may reflect the energy demand to coordinate multiple organs and contribute to cellular longevity.
Collapse
Affiliation(s)
- Ivo Vieira de Sousa Neto
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil.
| | - Ana Paula Pinto
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Vitor Rosetto Muñoz
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Rita de Cássia Marqueti
- Molecular Analysis Laboratory, Faculty of Ceilândia, Universidade de Brasília (UNB), Distrito Federal, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo 13484-350, Brazil
| | - Adelino Sanchez Ramos da Silva
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
4
|
Ioannidou A, Alatar S, Schipper R, Baganha F, Åhlander M, Hornell A, Fisher RM, Hagberg CE. Hypertrophied human adipocyte spheroids as in vitro model of weight gain and adipose tissue dysfunction. J Physiol 2021; 600:869-883. [PMID: 34387376 DOI: 10.1113/jp281445] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 08/09/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Adipocyte enlargement is a key feature of obesity and associated with insulin resistance and metabolic disease The cause and consequences of adipocyte enlargement have remained hard to study in vitro due to a lack of human cell models with representative morphology This paper provides an easily set up spheroid culture method, HUVAS (human unilocular vascularized adipocyte spheroids), for the differentiation and culturing of human adipocytes with a more unilocular morphology We show that providing adipocyte progenitors with a vascular differentiation niche is key for achieving in vitro differentiated adipocytes with large lipid droplets Lipid treatment of the HUVAS spheroids can further adipocyte enlargement and induce cellular dysfunction, mimicking the in vivo effects of weight gain The model will allow a wider research community to perform mechanistic studies of the factors impacting human adipocyte differentiation and growth, increasing our understanding of how obesity develops and why it has such detrimental consequences on whole body metabolism ABSTRACT: The rise in obesity prevalence has created an urgent need for new and improved methods to study human adipocytes and the pathogenic effects of weight gain in vitro. Despite the proven advantage of culturing adipocyte progenitors as 3D structures, the majority of studies continue using traditional 2D cultures which result in small, multilocular adipocytes with poor representability. We hypothesized that providing differentiating pre-adipocytes with a vascular growth niche would mimic in vivo adipogenesis and improve the differentiation into unilocular adipocytes. Here we present HUVAS (human unilocular vascularized adipocyte spheroids), a simple, easily applicable culture protocol that allows for the differentiation of human adipocytes with a more unilocular morphology and larger lipid droplets than previous protocols. Moreover, we offer a protocol for inducing adipocyte enlargement in vitro, resulting in larger lipid droplets and development of several key features of adipocyte dysfunction, including altered adipokine secretion, impaired lipolysis and insulin resistance. Taken together, our HUVAS model offers an improved culture system for studying the cellular and molecular mechanisms causing metabolic dysfunction and inflammation in human adipose tissue during weight gain. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Anna Ioannidou
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Shemim Alatar
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Ruby Schipper
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fabiana Baganha
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Matilda Åhlander
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Amanda Hornell
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Rachel M Fisher
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Carolina E Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Tharp WG, Gupta D, Sideleva O, Deacon CF, Holst JJ, Elahi D, Pratley RE. Effects of Pioglitazone on Glucose-Dependent Insulinotropic Polypeptide-Mediated Insulin Secretion and Adipocyte Receptor Expression in Patients With Type 2 Diabetes. Diabetes 2020; 69:146-157. [PMID: 31757794 DOI: 10.2337/db18-1163] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 11/13/2019] [Indexed: 11/13/2022]
Abstract
Incretin hormone dysregulation contributes to reduced insulin secretion and hyperglycemia in patients with type 2 diabetes mellitus (T2DM). Resistance to glucose-dependent insulinotropic polypeptide (GIP) action may occur through desensitization or downregulation of β-cell GIP receptors (GIP-R). Studies in rodents and cell lines show GIP-R expression can be regulated through peroxisome proliferator-activated receptor γ (PPARγ) response elements (PPREs). Whether this occurs in humans is unknown. To test this, we conducted a randomized, double-blind, placebo-controlled trial of pioglitazone therapy on GIP-mediated insulin secretion and adipocyte GIP-R expression in subjects with well-controlled T2DM. Insulin sensitivity improved, but the insulinotropic effect of infused GIP was unchanged following 12 weeks of pioglitazone treatment. In parallel, we observed increased GIP-R mRNA expression in subcutaneous abdominal adipocytes from subjects treated with pioglitazone. Treatment of cultured human adipocytes with troglitazone increased PPARγ binding to GIP-R PPREs. These results show PPARγ agonists regulate GIP-R expression through PPREs in human adipocytes, but suggest this mechanism is not important for regulation of the insulinotropic effect of GIP in subjects with T2DM. Because GIP has antilipolytic and lipogenic effects in adipocytes, the increased GIP-R expression may mediate accretion of fat in patients with T2DM treated with PPARγ agonists.
Collapse
Affiliation(s)
- William G Tharp
- Department of Anesthesiology, University of Vermont Medical Center, Larner College of Medicine, University of Vermont, Burlington, VT
| | - Dhananjay Gupta
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT
| | - Olga Sideleva
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT
| | - Carolyn F Deacon
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Cardiovascular Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Dariush Elahi
- Cardiovascular Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Richard E Pratley
- AdventHealth Translational Research Institute for Metabolism and Diabetes, Orlando, FL
| |
Collapse
|
6
|
Okumura T, Ohuchida K, Kibe S, Iwamoto C, Ando Y, Takesue S, Nakayama H, Abe T, Endo S, Koikawa K, Sada M, Horioka K, Mochidome N, Arita M, Moriyama T, Nakata K, Miyasaka Y, Ohtsuka T, Mizumoto K, Oda Y, Hashizume M, Nakamura M. Adipose tissue-derived stromal cells are sources of cancer-associated fibroblasts and enhance tumor progression by dense collagen matrix. Int J Cancer 2019; 144:1401-1413. [DOI: 10.1002/ijc.31775] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Takashi Okumura
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Shin Kibe
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Chika Iwamoto
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Yohei Ando
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Shin Takesue
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Hiromichi Nakayama
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Toshiya Abe
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Sho Endo
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Kazuhiro Koikawa
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Masafumi Sada
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Kohei Horioka
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Naoki Mochidome
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
- Department of Anatomic Pathology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Makoto Arita
- Department of Materials Science and Engineering, Faculty of Engineering; Kyushu University; Fukuoka Japan
| | - Taiki Moriyama
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Kohei Nakata
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Yoshihiro Miyasaka
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Takao Ohtsuka
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Kazuhiro Mizumoto
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Makoto Hashizume
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| |
Collapse
|
7
|
Abstract
During the last decades, research on adipose tissues has spread in parallel with the extension of obesity. Several observations converged on the idea that adipose tissues are organized in a large organ with endocrine and plastic properties. Two parenchymal components: white (WATs) and brown adipose tissues (BATs) are contained in subcutaneous and visceral compartments. Although both have endocrine properties, their function differs: WAT store lipids to allow intervals between meals, BAT burns lipids for thermogenesis. In spite of these opposite functions, they share the ability for reciprocal reversible transdifferentiation to tackle special physiologic needs. Thus, chronic need for thermogenesis induces browning and chronic positive energy balance induce whitening. Lineage tracing and data from explant studies strongly suggest other remodeling properties of this organ. During pregnancy and lactation breast WAT transdifferentiates into milk-secreting glands, composed by cells with abundant cytoplasmic lipids (pink adipocytes) and in the postlactation period pink adipocytes transdifferentiate back into WAT and BAT. The plastic properties of mature adipocytes are supported also by a liposecretion process in vitro where adult cell in culture transdifferentiate to differentiated fibroblast-like elements able to give rise to different phenotypes (rainbow adipocytes). In addition, the inflammasome system is activated in stressed adipocytes from obese adipose tissue. These adipocytes die and debris are reabsorbed by macrophages inducing a chronic low-grade inflammation, potentially contributing to insulin resistance and T2 diabetes. Thus, the plastic properties of this organ could open new therapeutic perspectives in the obesity-related metabolic disease and in breast pathologies. © 2018 American Physiological Society. Compr Physiol 8:1357-1431, 2018.
Collapse
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
8
|
Wang X, Xu S, Chen L, Shen D, Cao Y, Tang R, Wang X, Ji C, Li Y, Cui X, Guo X. Profiling Analysis Reveals the Potential Contribution of Peptides to Human Adipocyte Differentiation. Proteomics Clin Appl 2018; 12:e1700172. [PMID: 30009563 DOI: 10.1002/prca.201700172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 05/15/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE Peptide drugs provide promising regimes in anti-obesity treatment. In order to identify potential bioactive peptides involved in adipogenesis. EXPERIMENTAL DESIGN The intracellular peptides between preadipocytes and adipocytes are compared by liquid chromatography/mass spectrometry. The underlying biological function of the identified peptides are evaluated by gene ontology (GO) and pathway analysis of their precursors. To find more potential bioactive peptides, 50 peptide sequences are identified located in the functional domains with the use of the SMART and UniProt databases. Finally, the Open Targets Platform database is used to investigate the precursors related to metabolic diseases. RESULTS A total of 181 downregulated peptides and 89 upregulated peptides after differentiation (fold change > 1.5 and p-value < 0.05) are identified. The GO and pathway analysis indicate that these differentially expressed peptides play a role in adipogenesis. A total of 10 putative peptides 6 to 26 amino acids in length are identified that might have bioactive effects in adipogenesis and metabolic diseases. CONCLUSIONS AND CLINICAL RELEVANCE On one hand, present preliminary research provides a better understanding of the intracellular peptides during adipocyte differentiation. On the other hand, it lays a foundation for discovering new peptide drugs in anti-obesity treatment.
Collapse
Affiliation(s)
- Xingyun Wang
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Siliang Xu
- Center of Clinical Reproductive Medicine, the First Affiliated Hospital of Nanjing Medical University, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Ling Chen
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Dan Shen
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Yan Cao
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Ranran Tang
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Xing Wang
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Chenbo Ji
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Yun Li
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Xianwei Cui
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Xirong Guo
- Nanjing Maternity and Child Health Care Institute, Nanjing Maternity and Child Health Care Hospital, Obsterics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| |
Collapse
|
9
|
Nicu C, Pople J, Bonsell L, Bhogal R, Ansell DM, Paus R. A guide to studying human dermal adipocytes in situ. Exp Dermatol 2018; 27:589-602. [DOI: 10.1111/exd.13549] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Carina Nicu
- Centre for Dermatology Research; The University of Manchester; Manchester UK
- NIHR Manchester Biomedical Research Centre; Manchester Academic Health Science Centre; Manchester UK
| | | | - Laura Bonsell
- Centre for Dermatology Research; The University of Manchester; Manchester UK
- NIHR Manchester Biomedical Research Centre; Manchester Academic Health Science Centre; Manchester UK
| | | | - David M. Ansell
- Centre for Dermatology Research; The University of Manchester; Manchester UK
- NIHR Manchester Biomedical Research Centre; Manchester Academic Health Science Centre; Manchester UK
| | - Ralf Paus
- Centre for Dermatology Research; The University of Manchester; Manchester UK
- NIHR Manchester Biomedical Research Centre; Manchester Academic Health Science Centre; Manchester UK
- Department of Dermatology and Cutaneous Surgery; Miller School of Medicine; University of Miami; Miami FL USA
| |
Collapse
|
10
|
Zeng R, Fang Y, Zhang Y, Bai S. p62 is linked to mitophagy in oleic acid-induced adipogenesis in human adipose-derived stromal cells. Lipids Health Dis 2018; 17:133. [PMID: 29866118 PMCID: PMC5987550 DOI: 10.1186/s12944-018-0733-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/03/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Obesity is closely related to the abnormal differentiation of adipocytes, which are subjected to high plasma levels of free fatty acids (FFAs). As the most abundant FFA in the bloodstream, oleic acid (OA) has the ability to induce adipogenic differentiation in human adipose-derived stromal cells (hADSCs). Recently, p62, an autophagy mediator, has been shown to play a role in obesity and adipose tissue metabolism. Therefore, the aim of this study was to investigate the roles of autophagy and mitochondrial function at different stages of OA (in combination with insulin and dexamethasone)-induced adipogenesis in hADSCs. METHODS The hADSCs were incubated with OA, insulin, and dexamethasone after pretreatment with autophagy inhibitors or knockdown of p62 with shRNA. The adiposeness level was then analyzed by oil red O staining in the cells. The related proteins or mRNA levels were detected by western blot analysis or quantitative real-time polymerase chain reaction (PCR). RESULTS Treatment with 80 μM OA (substituted for isobutylmethylxantine; IBMX) for 10 days successfully induced hADSCs to adipocytes. During OA-induced adipogenesis, autophagy was induced, with an increased LC3II/I ratio on day 3 and a decreased protein level of p62 on and after day 3. Inhibition of autophagy with 3-methyladenine (3MA) at the early stage (day 0 to day 3) of differentiation, but not at the middle or late stage, significantly decreased OA-induced adipogenesis; while knockdown of p62 with shRNA significantly promoted adipogenesis in hADSCs. Moreover, the copy number of mtDNA (the ND1 gene) and the protein level of TOM20, a mitochondrial membrane protein, were increased following OA treatment, which was related to the stability of mitochondria. Interestingly, knockdown of p62 increased the mito-LC3II/I and cyto-LC3II/I ratios by 110.1% and 73.3%, respectively. The increase in the ratio of mito-LC3II/I was higher than that of cyto-LC3II/I. Furthermore, p62 knockdown-enhanced adipogenesis in hADSCs was abolished by inhibiting mitophagy with cyclosporine A. CONCLUSIONS These results suggested that p62 plays a protective role in adipogenesis of hADSCs through regulating mitophagy.
Collapse
Affiliation(s)
- Ruixia Zeng
- Department of Tissue Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110001, China
- Department of Anatomy, College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121001, China
- Liaoning Key Laboratory of Follicular Development and Reproductive Health, Jinzhou, 121001, China
| | - Yan Fang
- Department of Anatomy, College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121001, China
| | - Yibo Zhang
- Department of Pathogenic Biology, College of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, 121001, China
| | - Shuling Bai
- Department of Tissue Engineering, School of Fundamental Sciences, China Medical University, Shenyang, 110001, China.
| |
Collapse
|
11
|
Evaluation and optimization of differentiation conditions for human primary brown adipocytes. Sci Rep 2018; 8:5304. [PMID: 29593245 PMCID: PMC5871774 DOI: 10.1038/s41598-018-23700-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/19/2018] [Indexed: 12/15/2022] Open
Abstract
As an effective way to improve energy expenditure, increasing the mass and activity of brown adipose tissue (BAT) has become a promising treatment for obesity and its associated disorders. Many efforts have been made to promote brown adipogenesis and increase the thermogenic capacity of brown adipose cells (BACs). The present culture schemes for human BAC differentiation are mostly derived from white adipocyte differentiation schemes. To solve this issue, we compared the adipogenic and thermogenic effects of various components on human BAC differentiation and optimized their concentrations as well as the culture time for BAC differentiation. In this study, we found that the induction factors did not show a dose-dependent promotion of brown adipogenesis or thermogenic capacity. The higher differentiation levels did not inevitably result in higher BAT-specific gene expression levels or increased β3-receptor agonist sensitivity. As an important element of culture medium, triiodothyronine was found to be essential for differentiation and metabolic property maintenance. Furthermore, compared with other reported methods, this protocol induced a specific intrinsic differentiation program. Our study provides not only an optimized method for human BAC differentiation but also a cell model with good differentiation and thermogenic capacity for brown adipose research.
Collapse
|
12
|
Pu Y, Veiga-Lopez A. PPARγ agonist through the terminal differentiation phase is essential for adipogenic differentiation of fetal ovine preadipocytes. Cell Mol Biol Lett 2017; 22:6. [PMID: 28536637 PMCID: PMC5415806 DOI: 10.1186/s11658-017-0037-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 03/10/2017] [Indexed: 01/07/2023] Open
Abstract
Background Although the 3T3-L1 preadipocyte cell line represents an informative model for in vitro adipogenesis research, primary cultured cells are often needed to understand particular human or animal metabolic phenotypes. As demonstrated by in vitro cultured preadipocytes from large mammalian species, primary cultured cells require specific adipogenic differentiation conditions different to that of the 3T3-L1 cell line. These conditions are also species-specific and require optimization steps. However, efficient protocols to differentiate primary preadipocytes using alternative species to rodents are scarce. Sheep represent an amenable animal model for fetal biology and developmental origins of health and disease studies. In this work, we present with the first detailed procedure to efficiently differentiate primary fetal and adult ovine preadipocytes. Methods Fetal and adult ovine adipose and skin tissue harvest, preadipocyte and fibroblast isolation, proliferation, and standardization and optimization of a new adipogenic differentiation protocol. Use of commercial cell lines (3T3-L1 and NIH-3T3) for validation purposes. Oil red O stain and gene expression were used to validate adipogenic differentiation. ANOVA and Fisher’s exact test were used to determine statistical significance. Results Our optimized adipogenic differentiation method included a prolonged adipogenic cocktail exposure time from 2 to 8 days, higher insulin concentration, and supplementation with the peroxisome proliferator-activated receptor gamma (PPARγ) agonist, rosiglitazone. This protocol was optimized for both, fetal and adult preadipocytes. Conclusions Our protocol enables successful adipogenic differentiation of fetal and adult ovine preadipocytes. This work demonstrates that compared to the 3T3-L1 cell line, fetal ovine preadipocytes require a longer exposure to the differentiation cocktail, and the need for IMBX, dexamethasone, and/or the PPARγ agonist rosiglitazone through the terminal differentiation phase. They also require higher insulin concentration during differentiation to enhance lipid accumulation and similar to human primary preadipocytes, PPARγ agonist supplementation is also required for ovine adipogenic differentiation. This work highlights species-specific differences requirements for adipogenic differentiation and the need to develop standardized methods to investigate comparative adipocyte biology.
Collapse
Affiliation(s)
- Yong Pu
- Department of Animal Science, Michigan State University, 474 S. Shaw Lane Rm 1230 F, East Lansing, MI 48824 USA
| | - Almudena Veiga-Lopez
- Department of Animal Science, Michigan State University, 474 S. Shaw Lane Rm 1230 F, East Lansing, MI 48824 USA
| |
Collapse
|
13
|
Baker NA, Muir LA, Lumeng CN, O'Rourke RW. Differentiation and Metabolic Interrogation of Human Adipocytes. Methods Mol Biol 2017; 1566:61-76. [PMID: 28244041 DOI: 10.1007/978-1-4939-6820-6_7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Adipocytes differentiated from preadipocytes provide a valuable model for the study of human adipocyte metabolism. We describe methods for isolation of human stromal vascular cells, expansion of preadipocytes, differentiation into mature adipocytes, and in vitro metabolic interrogation of adipocytes.
Collapse
Affiliation(s)
- Nicki A Baker
- Section of General Surgery, Department of Surgery, University of Michigan Medical School, 2210 Taubman Center-5343, 1500 E. Medical Center Drive, Ann Arbor, MI, USA
| | - Lindsey A Muir
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Robert W O'Rourke
- Section of General Surgery, Department of Surgery, University of Michigan Medical School, 2210 Taubman Center-5343, 1500 E. Medical Center Drive, Ann Arbor, MI, USA. .,Department of Surgery, Ann Arbor Veteran's Administration Hospital, Ann Arbor, MI, USA.
| |
Collapse
|
14
|
Tharp WG, Gupta D, Smith J, Jones KP, Jones AM, Pratley RE. Effects of glucose and insulin on secretion of amyloid-β by human adipose tissue cells. Obesity (Silver Spring) 2016; 24:1471-9. [PMID: 27172814 PMCID: PMC5084817 DOI: 10.1002/oby.21494] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/05/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Obesity and type 2 diabetes mellitus are risk factors for developing Alzheimer disease. Overlapping patterns of metabolic dysfunction may be common molecular links between these complex diseases. Amyloid-β (Aβ) precursor protein and associated β- and γ-secretases are expressed in adipose tissue. Aβ precursor protein is up-regulated with obesity and correlated to insulin resistance. Aβ may be secreted by adipose tissue, its production may be regulated through metabolic pathways, and Aβ may exert effects on adipose tissue insulin receptor signaling. METHODS Human stromal-vascular cells and differentiated adipocytes were cultured with different combinations of glucose and insulin and then assayed for Aβ in conditioned media. Aβ was measured in vivo using adipose tissue microdialysis. RESULTS Aβ secretion was increased by glucose and insulin in vitro. Adipose tissue microdialysates contained Aβ. Adipocytes treated with Aβ had decreased expression of insulin receptor substrate-2 and reduced Akt-1 phosphorylation. CONCLUSIONS Aβ was made by adipose tissue cells in vitro at concentrations similar to in vivo measurements. Regulation of Aβ production by glucose and insulin and effects of Aβ on the insulin receptor pathway suggest similar cellular mechanisms may exist between neuronal dysfunction in Alzheimer disease and adipose dysfunction in type 2 diabetes.
Collapse
Affiliation(s)
- William G. Tharp
- Department of Medicine Endocrine UnitUniversity of Vermont College of MedicineBurlingtonVermontUSA
| | - Dhananjay Gupta
- Department of Medicine Endocrine UnitUniversity of Vermont College of MedicineBurlingtonVermontUSA
| | - Joshua Smith
- Florida Hospital Sanford/Burnham Translational Research Institute for Metabolism and DiabetesOrlandoFloridaUSA
| | - Karen P. Jones
- Florida Hospital Sanford/Burnham Translational Research Institute for Metabolism and DiabetesOrlandoFloridaUSA
| | - Amanda M. Jones
- Florida Hospital Sanford/Burnham Translational Research Institute for Metabolism and DiabetesOrlandoFloridaUSA
| | - Richard E. Pratley
- Florida Hospital Sanford/Burnham Translational Research Institute for Metabolism and DiabetesOrlandoFloridaUSA
| |
Collapse
|
15
|
Coimplanted endothelial cells improve adipose tissue grafts' survival by increasing vascularization. J Craniofac Surg 2016; 26:358-64. [PMID: 25675023 DOI: 10.1097/scs.0000000000001470] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND With goal of improving fat graft survival, many studies have focused on supplementing cells in the graft fat. In these studies, enhanced vascularization is considered the most important mechanism for the improved graft survival. Endothelial cells (ECs) are essential in vessel formation of the vascularization. Therefore, in this study, we coimplanted ECs with adipose tissue to investigate whether the ECs can enhance graft survival in a cell concentration-dependent manner. METHODS Endothelial cells were isolated from stromal vascular fraction derived from human liposuction aspirates, and the EC characteristics were confirmed by CD31 immunofluorescence staining, measuring acetylated low-density lipoprotein uptake, and observing the formation of capillary-like tubular structures in Matrigel. During the animal experiment, the isolated ECs were labeled, then added to 0.5-mL fat grafts at different numbers (0.5 × 10(6), 1 × 10(6), 2 × 10(6), and 4 × 10(6) cells) before subcutaneous implantation in nude mice. Grafts were harvested at 1 week, 1 month, and 2 months after -transplantation, and graft survival and vascularization were evaluated based on weight measurements, histological assessment, and vascular gene expression. RESULTS Stromal vascular fraction-derived vascular cells exhibited typical EC characteristics. The observed differences in explanted graft weight, vessel density, vascular gene expression, and cell tracking result indicated that coimplantation with ECs accelerated vascularization that increased graft survival in a concentration-dependent manner. Over the experimental period, fat grafts implanted with 4 × 10(6) ECs showed no weight loss and the greatest increases in measures of vascularization. CONCLUSIONS Endothelial cells can effectively enhance vascularization in fat grafts, and higher EC concentrations (eg, 4 × 10(6) ECs/0.5 mL adipose tissue) may best support graft survival.
Collapse
|
16
|
Biotin-dependent functions in adiposity: a study of monozygotic twin pairs. Int J Obes (Lond) 2015; 40:788-95. [PMID: 26601567 DOI: 10.1038/ijo.2015.237] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/16/2015] [Accepted: 11/06/2015] [Indexed: 12/28/2022]
|
17
|
Kang I, Okla M, Chung S. Ellagic acid inhibits adipocyte differentiation through coactivator-associated arginine methyltransferase 1-mediated chromatin modification. J Nutr Biochem 2014; 25:946-53. [DOI: 10.1016/j.jnutbio.2014.04.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/04/2014] [Accepted: 04/15/2014] [Indexed: 12/17/2022]
|
18
|
Lee MJ, Fried SK. Optimal protocol for the differentiation and metabolic analysis of human adipose stromal cells. Methods Enzymol 2014; 538:49-65. [PMID: 24529433 DOI: 10.1016/b978-0-12-800280-3.00004-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Obesity is reaching epidemic proportions so there is growing interest in the mechanisms that regulates adipose tissue development and function. Although murine adipose cell lines are useful for many mechanistic studies, primary human adipose stromal cells (ASCs), which can be isolated from distinct adipose depots and cultured in vitro, have clear translational relevance. We describe the methods to isolate, culture, and differentiate human ASCs to adipocytes that respond to physiologically relevant hormones, such as insulin and β-adrenergic agonists. We also describe methods for assaying hormonal effects on glucose transport and lipolysis.
Collapse
Affiliation(s)
- Mi-Jeong Lee
- Division of Endocrinology, Diabetes and Nutrition, Boston University School of Medicine, Boston, Massachusetts, USA.
| | - Susan K Fried
- Division of Endocrinology, Diabetes and Nutrition, Boston University School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
19
|
Abstract
Confirmation of the presence of functional brown adipose tissue (BAT) in humans has renewed interest in investigating the potential therapeutic use of this tissue. The finding that its activity positively correlates with decreased BMI, decreased fat content, and augmented energy expenditure suggests that increasing BAT mass/activity or browning of white adipose tissue (WAT) could be a strategy to prevent or treat obesity and its associated morbidities. The challenge now is to find a safe and efficient way to develop this idea. Whereas BAT has being widely studied in murine models both in vivo and in vitro, there is an urgent need for human cellular models to investigate BAT physiology and functionality from a molecular point of view. In this review, we focus on the latest insights surrounding BAT development and activation in rodents and humans. Then, we discuss how the availability of murine models has been essential to identify BAT progenitors and trace their lineage. Finally, we address how this information can be exploited to develop human cellular models for BAT differentiation/activation. In this context, human embryonic stem and induced pluripotent stem cells-based cellular models represent a resource of great potential value, as they can provide a virtually inexhaustible supply of starting material for functional genetic studies, -omics based analysis and validation of therapeutic approaches. Moreover, these cells can be readily genetically engineered, opening the possibility of generating patient-specific cellular models, allowing the investigation of the influence of different genetic backgrounds on BAT differentiation in pathological or in physiological states.
Collapse
Affiliation(s)
- Stefania Carobbio
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK Metabolic Research Laboratories, Addenbrooke's Treatment Centre, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
20
|
Zhao L, Ha JH, Okla M, Chung S. Activation of autophagy and AMPK by gamma-tocotrienol suppresses the adipogenesis in human adipose derived stem cells. Mol Nutr Food Res 2013; 58:569-79. [DOI: 10.1002/mnfr.201300157] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 07/24/2013] [Accepted: 08/03/2013] [Indexed: 12/27/2022]
Affiliation(s)
- Lu Zhao
- Department of Food Science and Human Nutrition; University of Florida; Gainesville FL USA
| | - Jung-Heun Ha
- Department of Food Science and Human Nutrition; University of Florida; Gainesville FL USA
| | - Meshail Okla
- Department of Food Science and Human Nutrition; University of Florida; Gainesville FL USA
| | - Soonkyu Chung
- Department of Food Science and Human Nutrition; University of Florida; Gainesville FL USA
| |
Collapse
|
21
|
White UA, Tchoukalova YD. Sex dimorphism and depot differences in adipose tissue function. Biochim Biophys Acta Mol Basis Dis 2013; 1842:377-92. [PMID: 23684841 DOI: 10.1016/j.bbadis.2013.05.006] [Citation(s) in RCA: 199] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 04/18/2013] [Accepted: 05/03/2013] [Indexed: 02/06/2023]
Abstract
Obesity, characterized by excessive adiposity, is a risk factor for many metabolic pathologies, such as type 2 diabetes mellitus (T2DM). Numerous studies have shown that adipose tissue distribution may be a greater predictor of metabolic health. Upper-body fat (visceral and subcutaneous abdominal) is commonly associated with the unfavorable complications of obesity, while lower-body fat (gluteal-femoral) may be protective. Current research investigations are focused on analyzing the metabolic properties of adipose tissue, in order to better understand the mechanisms that regulate fat distribution in both men and women. This review will highlight the adipose tissue depot- and sex-dependent differences in white adipose tissue function, including adipogenesis, adipose tissue developmental patterning, the storage and release of fatty acids, and secretory function. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
Affiliation(s)
- Ursula A White
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Yourka D Tchoukalova
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA.
| |
Collapse
|
22
|
Beranger GE, Karbiener M, Barquissau V, Pisani DF, Scheideler M, Langin D, Amri EZ. In vitro brown and “brite”/“beige” adipogenesis: Human cellular models and molecular aspects. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:905-14. [DOI: 10.1016/j.bbalip.2012.11.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Revised: 10/31/2012] [Accepted: 11/02/2012] [Indexed: 11/30/2022]
|
23
|
Ippoliti F, Canitano N, Businaro R. Stress and obesity as risk factors in cardiovascular diseases: a neuroimmune perspective. J Neuroimmune Pharmacol 2013; 8:212-26. [PMID: 23329173 DOI: 10.1007/s11481-012-9432-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 12/28/2012] [Indexed: 01/09/2023]
Abstract
Obesity is now growing at an alarming rate reaching epidemic proportions worldwide thus increasing morbidity and mortality rates for chronic disease. But although we have ample information on the complications associated with obesity, precisely what causes obesity remains poorly understood. Some evidence attributes a major role to a low-grade chronic inflammatory state (neurogenic inflammation) induced in obesity by inflammatory mediators produced and secreted within the expanded activated adipocyte pool. Adipose tissue is an endocrine organ that secretes numerous adipose tissue-specific or enriched hormones, known as adipokines, cytokine-like molecules thought to play a pathogenic role in cardiovascular diseases. The imbalance between increased inflammatory stimuli and decreased anti-inflammatory mechanisms may depend on chronic stress. Hence the positive correlation found between stress, obesity and cardiovascular diseases. The chronic inflammatory state associated with insulin resistance and endothelial dysfunction is highly deleterious for vascular function. This review focuses on the proposed neuroimmunodulatory mechanisms linking chronic (psychological) stress, obesity and cardiovascular diseases.
Collapse
Affiliation(s)
- Flora Ippoliti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy.
| | | | | |
Collapse
|
24
|
Sorisky A, Molgat ASD, Gagnon A. Macrophage-induced adipose tissue dysfunction and the preadipocyte: should I stay (and differentiate) or should I go? Adv Nutr 2013; 4:67-75. [PMID: 23319125 PMCID: PMC3648741 DOI: 10.3945/an.112.003020] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue can be regarded as a multidepot organ responsible for metabolic homeostasis by managing sophisticated energy transactions as well as by producing bioactive molecules that regulate insulin sensitivity and immune and vascular responses. Chronic nutrient excess expands adipose tissue, and concomitant variations in its cellular and matrix remodeling can affect the extent of the metabolic dysfunction that is associated with obesity. Preadipocytes, also termed adipose progenitor cells, play a pivotal role in determining whether a dysfunctional hypertrophic state arises as opposed to a hyperplastic process in which mature adipocytes remain relatively responsive. Obesity is associated with infiltration of macrophages, and these immune cells have been shown to communicate with preadipocytes to influence how they differentiate, survive, and proliferate. Understanding macrophage-preadipocyte interactions and their effect on adipose remodeling mechanisms may identify potential therapeutic molecular targets to improve adipose tissue function, even in the face of obesity.
Collapse
Affiliation(s)
- Alexander Sorisky
- Chronic Disease Program, Ottawa Hospital Research Institute, Ontario, Canada.
| | | | | |
Collapse
|
25
|
Lee MJ, Wu Y, Fried SK. A modified protocol to maximize differentiation of human preadipocytes and improve metabolic phenotypes. Obesity (Silver Spring) 2012; 20:2334-40. [PMID: 22627913 PMCID: PMC4320940 DOI: 10.1038/oby.2012.116] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Adipose stromal cells proliferate and differentiate into adipocytes, providing a valuable model system for studies of adipocyte biology. We compared differentiation protocols for human preadipocytes and report on their metabolic phenotypes. By simply prolonging the adipogenic induction period from the first 3 to 7 days, the proportion of cells acquiring adipocyte morphology increased from 30-70% to over 80% in human subcutaneous preadipocytes (passages 5-6). These morphological changes were accompanied by increases in the adipogenic marker expression and improved adipocyte metabolic phenotypes: enhanced responses to β-adrenergically stimulated lipolysis and to insulin-stimulated glucose metabolism into triglyceride (TG). Confirming previous studies, fetal bovine serum (FBS) dose-dependently inhibited adipogenesis. However, in subcutaneous preadipocytes that differentiate well (donor-dependant high capacity and subcultured fewer than two times), the use of 7d-induction protocols in both 3% FBS and serum-free conditions allowed >80% differentiation. Responsiveness to β-adrenergically stimulated lipolysis was lower in 3% FBS. Rates of insulin-stimulated glucose uptake were higher in adipocytes differentiated with 3% FBS, whereas the sensitivity to insulin was almost identical between the two groups. In summary, extending the length of the induction period in adipogenic cocktail improves the degree of differentiation and responses to key metabolic hormones. This protocol permits functional analysis of metabolic phenotypes in valuable primary human adipocyte cultures through multiple passages.
Collapse
Affiliation(s)
- Mi-Jeong Lee
- Correspondence to Mi-Jeong Lee PhD, Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, Boston University, Boston Nutrition and Obesity Research Center, 650 Albany St, EBRC-810, Boston, MA 02118 Tel: 617-638-8258; Fax: 617-638-7124;
| | | | | |
Collapse
|