1
|
Marjot T, Armstrong MJ, Stine JG. Skeletal muscle and MASLD: Mechanistic and clinical insights. Hepatol Commun 2025; 9:e0711. [PMID: 40408301 DOI: 10.1097/hc9.0000000000000711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 05/25/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is intrinsically linked with widespread metabolic perturbations, including within skeletal muscle. Indeed, MASLD is associated with a range of skeletal muscle abnormalities, including insulin resistance, myosteatosis, and sarcopenia, which all converge on the liver to drive disease progression and adverse patient outcomes. This review explores the mechanistic links between skeletal muscle and MASLD, including the role of abnormal glycemic control, systemic inflammation, and disordered myokine signaling. In turn, we discuss how intrinsic liver pathology can feed back to further exacerbate poor skeletal muscle health. Given the central importance of skeletal muscle in MASLD pathogenesis, it offers clinicians an opportunity to intervene for therapeutic benefit. We, therefore, summarize the role of nutrition and physical activity on skeletal muscle mass, quality, and metabolic function and discuss the knock-on effect this has on the liver. An awareness of these treatment strategies is particularly important in the era of effective pharmacological and surgical weight loss interventions, which can be associated with the development of sarcopenia. Finally, we highlight a number of promising drug agents in the clinical trial pipeline that specifically target skeletal muscle in an attempt to improve metabolic and physical functioning.
Collapse
Affiliation(s)
- Thomas Marjot
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), Radcliffe Department of Medicine, Churchill Hospital, University of Oxford, Oxford, UK
- Translational Gastroenterology and Liver Unit (TGLU), Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Matthew J Armstrong
- Liver Unit, Queen Elizabeth University Hospital Birmingham, Birmingham, UK
- Birmingham NIHR Biomedical Research Centre, University of Birmingham, Birmingham, UK
| | - Jonathan G Stine
- Department of Medicine, Division of Gastroenterology and Hepatology, Penn State Health-Milton S. Hershey Medical Centre, Hershey, Pennsylvania, USA
| |
Collapse
|
2
|
Cuesta-Margolles G, Schlecht-Louf G, Bachelerie F. ACKR3 in Skin Homeostasis, an Overlooked Player in the CXCR4/CXCL12 Axis. J Invest Dermatol 2025; 145:1039-1049. [PMID: 39466217 DOI: 10.1016/j.jid.2024.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 10/29/2024]
Abstract
CXCL12 and its receptor CXCR4 emerge as critical regulators within the intricate network of processes ensuring skin homeostasis. In this review, we discuss their spatial distribution and function in steady-state skin; delve into their role in acute wound healing, with emphasis on fibrotic and regenerative responses; and explore their relevance in skin responses to commensals and pathogens. Given the lack of knowledge surrounding ACKR3, the atypical receptor of CXCL12, we speculate whether and how it might be involved in the processes mentioned earlier. Is ACKR3 the (a)typical friend who enjoys missing the party, or do we need to take a closer look?
Collapse
Affiliation(s)
| | - Géraldine Schlecht-Louf
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Françoise Bachelerie
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| |
Collapse
|
3
|
Zheng H, Wang C, Zhou A, Chen X. Transcriptomic and Lipidomic Characteristics of Subcutaneous Fat Deposition in Small-Sized Meat Ducks. Metabolites 2025; 15:158. [PMID: 40137123 PMCID: PMC11944229 DOI: 10.3390/metabo15030158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/15/2025] [Accepted: 02/20/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Subcutaneous fat deposition is associated with ducks' meat quality and the methods used to cook them. However, the reasons underlying the differences in the lipid deposition of small-sized Wuqin10 meat ducks remain unclear. Method: In the present study, to elucidate the metabolic mechanisms of lipid deposition, we comprehensively analyzed the transcriptomics and lipidomics of subcutaneous fat in Wuqin10 meat ducks with different subcutaneous thicknesses with six replicates. Results: A total of 1120 lipids were detected in the lipidomic analysis, and 39 lipids were inexorably regulated in the ducks with the thick subcutaneous layer compared to those with the thin layer; further, the up-regulated lipids were primarily triglycerides (TGs), which may have resulted in adipocyte enlargement. Furthermore, the transcriptomic analysis identified 265 differentially expressed genes (DEGs), including 119 down-regulated and 146 up-regulated genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed that the DEGs were significantly enriched in the histidine, arginine, proline metabolism signaling and adipocytokine signaling pathways. The protein-protein interaction (PPI) network in Cytoscape 3.8.2 identified hub genes HSP90AA1, RUNX2, ACTN2, ACTA1, IL10, CXCR4, EGF, SOCS3 and PTK2, which were associated with the JAK-STAT signaling pathway and regulation of adipocyte hypertrophy. Conclusion: Taken together, our findings reveal the patterns of lipids and the gene expression of subcutaneous fat, providing a basis for future studies of subcutaneous fat deposition in small-sized meat ducks.
Collapse
Affiliation(s)
- Hao Zheng
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China;
| | - Cui Wang
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China;
| | - Ao Zhou
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan 430023, China;
| | - Xing Chen
- Institute of Animal Husbandry and Veterinary, Wuhan Academy of Agricultural Science, Wuhan 430345, China
| |
Collapse
|
4
|
Liu J, Qi L, Bao S, Yan F, Chen J, Yu S, Dong C. The acute spinal cord injury microenvironment and its impact on the homing of mesenchymal stem cells. Exp Neurol 2024; 373:114682. [PMID: 38199509 DOI: 10.1016/j.expneurol.2024.114682] [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: 11/07/2023] [Revised: 12/08/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
Spinal cord injury (SCI) is a highly debilitating condition that inflicts devastating harm on the lives of affected individuals, underscoring the urgent need for effective treatments. By activating inflammatory cells and releasing inflammatory factors, the secondary injury response creates an inflammatory microenvironment that ultimately determines whether neurons will undergo necrosis or regeneration. In recent years, mesenchymal stem cells (MSCs) have garnered increasing attention for their therapeutic potential in SCI. MSCs not only possess multipotent differentiation capabilities but also have homing abilities, making them valuable as carriers and mediators of therapeutic agents. The inflammatory microenvironment induced by SCI recruits MSCs to the site of injury through the release of various cytokines, chemokines, adhesion molecules, and enzymes. However, this mechanism has not been previously reported. Thus, a comprehensive exploration of the molecular mechanisms and cellular behaviors underlying the interplay between the inflammatory microenvironment and MSC homing is crucial. Such insights have the potential to provide a better understanding of how to harness the therapeutic potential of MSCs in treating inflammatory diseases and facilitating injury repair. This review aims to delve into the formation of the inflammatory microenvironment and how it influences the homing of MSCs.
Collapse
Affiliation(s)
- Jinyi Liu
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Longju Qi
- Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Shengzhe Bao
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Fangsu Yan
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Jiaxi Chen
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Shumin Yu
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Chuanming Dong
- Department of Anatomy, Medical College of Nantong University, Nantong, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China.
| |
Collapse
|
5
|
Mathieu M, Girousse A, Sengenès C. [What if the origin of FAPs was contributing to their heterogeneity in muscle?]. Med Sci (Paris) 2023; 39 Hors série n° 1:15-21. [PMID: 37975765 DOI: 10.1051/medsci/2023129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023] Open
Abstract
Fibro-adipogenic progenitors (FAPs) are resident mesenchymal stromal cells (MSCs) of skeletal muscle. They play a crucial role in muscle homeostasis and regeneration through their paracrine activity. Recent technological advances in single-cell RNA sequencing have allowed the characterization of the heterogeneity within this cell population. In this article, we will present the different subpopulations of FAPs under basal, injury, or degenerative conditions, as well as their associated functions in mice and humans. We will then discuss the potential extramuscular origin of a post-injury FAP population. Indeed, our recent work demonstrates that MSCs from adipose tissue, infiltrating the muscle, could contribute to FAP heterogeneity.
Collapse
Affiliation(s)
- Maxime Mathieu
- Institut RESTORE, UMR Inserm 1301 / CNRS 5070, Toulouse, France
| | | | | |
Collapse
|
6
|
Zhang T, Li J, Li X, Liu Y. Intermuscular adipose tissue in obesity and related disorders: cellular origins, biological characteristics and regulatory mechanisms. Front Endocrinol (Lausanne) 2023; 14:1280853. [PMID: 37920255 PMCID: PMC10619759 DOI: 10.3389/fendo.2023.1280853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/01/2023] [Indexed: 11/04/2023] Open
Abstract
Intermuscular adipose tissue (IMAT) is a unique adipose depot interspersed between muscle fibers (myofibers) or muscle groups. Numerous studies have shown that IMAT is strongly associated with insulin resistance and muscular dysfunction in people with metabolic disease, such as obesity and type 2 diabetes. Moreover, IMAT aggravates obesity-related muscle metabolism disorders via secretory factors. Interestingly, researchers have discovered that intermuscular brown adipocytes in rodent models provide new hope for obesity treatment by acting on energy dissipation, which inspired researchers to explore the underlying regulation of IMAT formation. However, the molecular and cellular properties and regulatory processes of IMAT remain debated. Previous studies have suggested that muscle-derived stem/progenitor cells and other adipose tissue progenitors contribute to the development of IMAT. Adipocytes within IMAT exhibit features that are similar to either white adipocytes or uncoupling protein 1 (UCP1)-positive brown adipocytes. Additionally, given the heterogeneity of skeletal muscle, which comprises myofibers, satellite cells, and resident mesenchymal progenitors, it is plausible that interplay between these cellular components actively participate in the regulation of intermuscular adipogenesis. In this context, we review recent studies associated with IMAT to offer insights into the cellular origins, biological properties, and regulatory mechanisms of IMAT. Our aim is to provide novel ideas for the therapeutic strategy of IMAT and the development of new drugs targeting IMAT-related metabolic diseases.
Collapse
Affiliation(s)
- Ting Zhang
- Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
- Medical Research Center, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
| | - Jun Li
- Department of Orthopedics, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
| | - Xi Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Yanjun Liu
- Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
| |
Collapse
|
7
|
Bonora BM, Cappellari R, Albiero M, Prevedello L, Foletto M, Vettor R, Avogaro A, Fadini GP. Putative circulating adipose tissue-derived stem cells, obesity, and metabolic syndrome features. J Endocrinol Invest 2023; 46:2147-2155. [PMID: 36952215 PMCID: PMC10514150 DOI: 10.1007/s40618-023-02067-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/09/2023] [Indexed: 03/24/2023]
Abstract
PURPOSE In mice, adipose tissue-derived stem cells (ASCs) reach the systemic circulation and establish ectopic adipose depots fostering insulin resistance, but whether this occurs in humans is unknown. We examined circulating ASCs in individuals with various combination of metabolic syndrome traits. METHODS We enrolled patients attending a routine metabolic evaluation or scheduled for bariatric surgery. We quantified ASCs as CD34+CD45-CD31-(CD36+) cells in the stromal vascular fraction of subcutaneous and visceral adipose tissue samples and examined the presence and frequency of putative ASCs in peripheral blood. RESULTS We included 111 patients (mean age 59 years, 55% males), 40 of whom were scheduled for bariatric surgery. The population of CD34+CD45-CD31- ASCs was significantly more frequent in visceral than subcutaneous adipose depots (10.4 vs 4.1% of the stromal vascular fraction; p < 0.001), but not correlated with BMI or metabolic syndrome traits. The same phenotype of ASCs was detectable in peripheral blood of 58.6% of patients. Those with detectable circulating ASCs had significantly higher BMI (37.8 vs 33.3 kg/m2; p = 0.003) and waist (111.2 vs 105.4 cm; p = 0.001), but no difference in other metabolic syndrome traits (p = 0.84). After bariatric surgery, patients with detectable circulating ASCs had greater BMI reductions at 6 months (- 10.4 vs - 7.8 kg/m2; p = 0.014). CONCLUSION Presence of putative circulating ASCs, antigenically similar to those observed in the adipose tissue, is associated with greater adiposity and larger BMI reduction after surgery, but not with clinical signs of metabolic impairment. The role of circulating ASCs in adipose tissue biology and systemic metabolism deserves further investigation.
Collapse
Affiliation(s)
- B M Bonora
- Department of Medicine, University of Padova, 35128, Padua, Italy
- Veneto Institute of Molecular Medicine, 35129, Padua, Italy
| | - R Cappellari
- Department of Medicine, University of Padova, 35128, Padua, Italy
- Bariatric Surgery Unit, University Hospital of Padova, 35128, Padua, Italy
| | - M Albiero
- Department of Medicine, University of Padova, 35128, Padua, Italy
- Veneto Institute of Molecular Medicine, 35129, Padua, Italy
| | - L Prevedello
- Bariatric Surgery Unit, University Hospital of Padova, 35128, Padua, Italy
| | - M Foletto
- Bariatric Surgery Unit, University Hospital of Padova, 35128, Padua, Italy
| | - R Vettor
- Department of Medicine, University of Padova, 35128, Padua, Italy
| | - A Avogaro
- Department of Medicine, University of Padova, 35128, Padua, Italy
| | - G P Fadini
- Department of Medicine, University of Padova, 35128, Padua, Italy.
- Veneto Institute of Molecular Medicine, 35129, Padua, Italy.
| |
Collapse
|
8
|
Ghonim MA, Boyd DF, Flerlage T, Thomas PG. Pulmonary inflammation and fibroblast immunoregulation: from bench to bedside. J Clin Invest 2023; 133:e170499. [PMID: 37655660 PMCID: PMC10471178 DOI: 10.1172/jci170499] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
In recent years, there has been an explosion of interest in how fibroblasts initiate, sustain, and resolve inflammation across disease states. Fibroblasts contain heterogeneous subsets with diverse functionality. The phenotypes of these populations vary depending on their spatial distribution within the tissue and the immunopathologic cues contributing to disease progression. In addition to their roles in structurally supporting organs and remodeling tissue, fibroblasts mediate critical interactions with diverse immune cells. These interactions have important implications for defining mechanisms of disease and identifying potential therapeutic targets. Fibroblasts in the respiratory tract, in particular, determine the severity and outcome of numerous acute and chronic lung diseases, including asthma, chronic obstructive pulmonary disease, acute respiratory distress syndrome, and idiopathic pulmonary fibrosis. Here, we review recent studies defining the spatiotemporal identity of the lung-derived fibroblasts and the mechanisms by which these subsets regulate immune responses to insult exposures and highlight past, current, and future therapeutic targets with relevance to fibroblast biology in the context of acute and chronic human respiratory diseases. This perspective highlights the importance of tissue context in defining fibroblast-immune crosstalk and paves the way for identifying therapeutic approaches to benefit patients with acute and chronic pulmonary disorders.
Collapse
Affiliation(s)
- Mohamed A. Ghonim
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al Azhar University, Cairo, Egypt
| | - David F. Boyd
- Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, USA
| | - Tim Flerlage
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
9
|
Sastourné-Arrey Q, Mathieu M, Contreras X, Monferran S, Bourlier V, Gil-Ortega M, Murphy E, Laurens C, Varin A, Guissard C, Barreau C, André M, Juin N, Marquès M, Chaput B, Moro C, O'Gorman D, Casteilla L, Girousse A, Sengenès C. Adipose tissue is a source of regenerative cells that augment the repair of skeletal muscle after injury. Nat Commun 2023; 14:80. [PMID: 36604419 PMCID: PMC9816314 DOI: 10.1038/s41467-022-35524-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/08/2022] [Indexed: 01/07/2023] Open
Abstract
Fibro-adipogenic progenitors (FAPs) play a crucial role in skeletal muscle regeneration, as they generate a favorable niche that allows satellite cells to perform efficient muscle regeneration. After muscle injury, FAP content increases rapidly within the injured muscle, the origin of which has been attributed to their proliferation within the muscle itself. However, recent single-cell RNAseq approaches have revealed phenotype and functional heterogeneity in FAPs, raising the question of how this differentiation of regenerative subtypes occurs. Here we report that FAP-like cells residing in subcutaneous adipose tissue (ScAT), the adipose stromal cells (ASCs), are rapidly released from ScAT in response to muscle injury. Additionally, we find that released ASCs infiltrate the damaged muscle, via a platelet-dependent mechanism and thus contribute to the FAP heterogeneity. Moreover, we show that either blocking ASCs infiltration or removing ASCs tissue source impair muscle regeneration. Collectively, our data reveal that ScAT is an unsuspected physiological reservoir of regenerative cells that support skeletal muscle regeneration, underlining a beneficial relationship between muscle and fat.
Collapse
Affiliation(s)
- Quentin Sastourné-Arrey
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Maxime Mathieu
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Xavier Contreras
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Sylvie Monferran
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Virginie Bourlier
- Institute of Metabolic and Cardiovascular Diseases, INSERM /Paul Sabatier University UMR 1297, Team MetaDiab, Toulouse, France
| | - Marta Gil-Ortega
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Enda Murphy
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
| | - Claire Laurens
- Institute of Metabolic and Cardiovascular Diseases, INSERM /Paul Sabatier University UMR 1297, Team MetaDiab, Toulouse, France
| | - Audrey Varin
- RESTORE, Research Center, Team 2 FLAMES, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Christophe Guissard
- RESTORE, Research Center, Team 4 GOT-IT, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Corinne Barreau
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Mireille André
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Noémie Juin
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Marie Marquès
- Institute of Metabolic and Cardiovascular Diseases, INSERM /Paul Sabatier University UMR 1297, Team MetaDiab, Toulouse, France
| | - Benoit Chaput
- Department of Plastic and Reconstructive Surgery, Toulouse University Hospital, 31100, Toulouse, France
| | - Cédric Moro
- Institute of Metabolic and Cardiovascular Diseases, INSERM /Paul Sabatier University UMR 1297, Team MetaDiab, Toulouse, France
| | - Donal O'Gorman
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
| | - Louis Casteilla
- RESTORE, Research Center, Team 4 GOT-IT, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Amandine Girousse
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Coralie Sengenès
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France.
| |
Collapse
|
10
|
Goodpaster BH, Bergman BC, Brennan AM, Sparks LM. Intermuscular adipose tissue in metabolic disease. Nat Rev Endocrinol 2022; 19:285-298. [PMID: 36564490 DOI: 10.1038/s41574-022-00784-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2022] [Indexed: 12/24/2022]
Abstract
Intermuscular adipose tissue (IMAT) is a distinct adipose depot described in early reports as a 'fatty replacement' or 'muscle fat infiltration' that was linked to ageing and neuromuscular disease. Later studies quantifying IMAT with modern in vivo imaging methods (computed tomography and magnetic resonance imaging) revealed that IMAT is proportionately higher in men and women with type 2 diabetes mellitus and the metabolic syndrome than in people without these conditions and is associated with insulin resistance and poor physical function with ageing. In parallel, agricultural research has provided extensive insight into the role of IMAT and other muscle lipids in muscle (that is, meat) quality. In addition, studies using rodent models have shown that IMAT is a bona fide white adipose tissue depot capable of robust triglyceride storage and turnover. Insight into the importance of IMAT in human biology has been limited by the dearth of studies on its biological properties, that is, the quality of IMAT. However, in the past few years, investigations have begun to determine that IMAT has molecular and metabolic features that distinguish it from other adipose tissue depots. These studies will be critical to further decipher the role of IMAT in health and disease and to better understand its potential as a therapeutic target.
Collapse
Affiliation(s)
| | - Bryan C Bergman
- Division of Endocrinology, Diabetes, and Metabolism, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrea M Brennan
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| |
Collapse
|
11
|
Plaza A, Merino B, Ruiz-Gayo M. Cholecystokinin promotes functional expression of the aquaglycerol channel aquaporin 7 in adipocytes. Br J Pharmacol 2022; 179:4092-4106. [PMID: 35366004 DOI: 10.1111/bph.15848] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 03/16/2022] [Accepted: 03/20/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Cholecystokinin (CCK) promotes triglyceride storage and adiponectin production in white adipose tissue (WAT), suggesting that CCK modulates WAT homeostasis. Our goal was to investigate the role of CCK in regulating the expression and function of the aquaglycerol channel aquaporin 7 (AQP7), a protein that is pivotal for maintaining adipocyte homeostasis and preserving insulin responsiveness. EXPERIMENTAL APPROACH The effect of the bioactive fragment of CCK, CCK-8, in regulating adipose AQP7 expression and glycerol efflux was assessed in rats as well as in pre-adipocytes. Moreover, the involvement of insulin receptors in the effects of CCK-8 was characterized in pre-adipocytes lacking insulin receptors. KEY RESULTS CCK-8 induced AQP7 gene expression in rat WAT, concomitantly increasing plasma glycerol concentration. In isolated pre-adipocytes, CCK-8 also enhanced both AQP7 expression and glycerol leakage. The effect of CCK-8 was independent of the lipolysis rate, as CCK-8 failed to promote fatty acid release by adipocytes. In addition, CCK-8 did not enhance hormone sensitive lipase phosphorylation, which is the rate-limiting step of lipolysis. Moreover, the effects of CCK-8 were dependent on the activation of protein kinase B and PPARγ. Silencing insulin receptor (IR) expression inhibited CCK-8-induced Aqp7 expression in pre-adipocytes. Furthermore, insulin enhanceded the effect of CCK-8. CONCLUSIONS AND IMPLICATIONS CCK regulates AQP7 expression and function, and this effect is dependent on insulin. Accordingly, CCK receptor agonists could be suitable for preserving and improving insulin responsiveness in WAT.
Collapse
Affiliation(s)
- Adrián Plaza
- Departamento de Ciencias Farmacéuticas y de la Salud. Facultad de Farmacia. Universidad CEU - San Pablo. CEU Universities, Madrid, Spain.,Laboratory of Bioactive Products and Metabolic Syndrome, IMDEA Food Institute, Madrid, Spain
| | - Beatriz Merino
- Departamento de Ciencias Farmacéuticas y de la Salud. Facultad de Farmacia. Universidad CEU - San Pablo. CEU Universities, Madrid, Spain
| | - Mariano Ruiz-Gayo
- Departamento de Ciencias Farmacéuticas y de la Salud. Facultad de Farmacia. Universidad CEU - San Pablo. CEU Universities, Madrid, Spain
| |
Collapse
|
12
|
Favaretto F, Bettini S, Busetto L, Milan G, Vettor R. Adipogenic progenitors in different organs: Pathophysiological implications. Rev Endocr Metab Disord 2022; 23:71-85. [PMID: 34716543 PMCID: PMC8873140 DOI: 10.1007/s11154-021-09686-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 12/14/2022]
Abstract
In physiological conditions, the adipose organ resides in well-defined areas, where it acts providing an energy supply and as an endocrine organ involved in the control of whole-body energy metabolism. Adipose tissue adipokines connect the body's nutritional status to the regulation of energy balance. When it surrounds organs, it provides also for mechanical protection. Adipose tissue has a complex and heterogenous cellular composition that includes adipocytes, adipose tissue-derived stromal and stem cells (ASCs) which are mesenchymal stromal cells, and endothelial and immune cells, which signal to each other and to other tissues to maintain homeostasis. In obesity and in other nutrition related diseases, as well as in age-related diseases, biological and functional changes of adipose tissue give rise to several complications. Obesity triggers alterations of ASCs, impairing adipose tissue remodeling and adipose tissue function, which induces low-grade systemic inflammation, progressive insulin resistance and other metabolic disorders. Adipose tissue grows by hyperplasia recruiting new ASCs and by hypertrophy, up to its expandability limit. To overcome this limitation and to store the excess of nutrients, adipose tissue develops ectopically, involving organs such as muscle, bone marrow and the heart. The origin of ectopic adipose organ is not clearly elucidated, and a possible explanation lies in the stimulation of the adipogenic differentiation of mesenchymal precursor cells which normally differentiate toward a lineage specific for the organ in which they reside. The chronic exposition of these newly-formed adipose depots to the pathological environment, will confer to them all the phenotypic characteristics of a dysfunctional adipose tissue, perpetuating the organ alterations. Visceral fat, but also ectopic fat, either in the liver, muscle or heart, can increase the risk of developing insulin resistance, type 2 diabetes, and cardiovascular diseases. Being able to prevent and to target dysfunctional adipose tissue will avoid the progression towards the complications of obesity and other nutrition-related diseases. The aim of this review is to summarize some of the knowledge regarding the presence of adipose tissue in particular tissues (where it is not usually present), describing the composition of its adipogenic precursors, and the interactions responsible for the development of organ pathologies.
Collapse
Affiliation(s)
- Francesca Favaretto
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Silvia Bettini
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Luca Busetto
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Gabriella Milan
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| | - Roberto Vettor
- grid.5608.b0000 0004 1757 3470Department of Medicine, Internal Medicine 3, University of Padua, via Giustiniani 2, 35128 Padua, Italy
| |
Collapse
|
13
|
Makris S, de Winde CM, Horsnell HL, Cantoral-Rebordinos JA, Finlay RE, Acton SE. Immune function and dysfunction are determined by lymphoid tissue efficacy. Dis Model Mech 2022; 15:dmm049256. [PMID: 35072206 PMCID: PMC8807573 DOI: 10.1242/dmm.049256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Lymphoid tissue returns to a steady state once each immune response is resolved, and although this occurs multiple times throughout life, its structural integrity and functionality remain unaffected. Stromal cells orchestrate cellular interactions within lymphoid tissue, and any changes to the microenvironment can have detrimental outcomes and drive disease. A breakdown in lymphoid tissue homeostasis can lead to a loss of tissue structure and function that can cause aberrant immune responses. This Review highlights recent advances in our understanding of lymphoid tissue function and remodelling in adaptive immunity and in disease states. We discuss the functional role of lymphoid tissue in disease progression and explore the changes to lymphoid tissue structure and function driven by infection, chronic inflammatory conditions and cancer. Understanding the role of lymphoid tissues in immune responses to a wide range of pathologies allows us to take a fuller systemic view of disease progression.
Collapse
Affiliation(s)
- Spyridon Makris
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Charlotte M. de Winde
- Department for Molecular Cell Biology and Immunology, Amsterdam UMC, location VUmc, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands
| | - Harry L. Horsnell
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jesús A. Cantoral-Rebordinos
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Rachel E. Finlay
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester M13 9PL, UK
| | - Sophie E. Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
14
|
Markov A, Thangavelu L, Aravindhan S, Zekiy AO, Jarahian M, Chartrand MS, Pathak Y, Marofi F, Shamlou S, Hassanzadeh A. Mesenchymal stem/stromal cells as a valuable source for the treatment of immune-mediated disorders. Stem Cell Res Ther 2021; 12:192. [PMID: 33736695 PMCID: PMC7971361 DOI: 10.1186/s13287-021-02265-1] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Over recent years, mesenchymal stem/stromal cells (MSCs) and their potential biomedical applications have received much attention from the global scientific community in an increasing manner. Firstly, MSCs were successfully isolated from human bone marrow (BM), but in the next steps, they were also extracted from other sources, mostly from the umbilical cord (UC) and adipose tissue (AT). The International Society for Cellular Therapy (ISCT) has suggested minimum criteria to identify and characterize MSCs as follows: plastic adherence, surface expression of CD73, D90, CD105 in the lack of expression of CD14, CD34, CD45, and human leucocyte antigen-DR (HLA-DR), and also the capability to differentiate to multiple cell types including adipocyte, chondrocyte, or osteoblast in vitro depends on culture conditions. However, these distinct properties, including self-renewability, multipotency, and easy accessibility are just one side of the coin; another side is their huge secretome which is comprised of hundreds of mediators, cytokines, and signaling molecules and can effectively modulate the inflammatory responses and control the infiltration process that finally leads to a regulated tissue repair/healing or regeneration process. MSC-mediated immunomodulation is a direct result of a harmonic synergy of MSC-released signaling molecules (i.e., mediators, cytokines, and chemokines), the reaction of immune cells and other target cells to those molecules, and also feedback in the MSC-molecule-target cell axis. These features make MSCs a respectable and eligible therapeutic candidate to be evaluated in immune-mediated disorders, such as graft versus host diseases (GVHD), multiple sclerosis (MS), Crohn's disease (CD), and osteoarthritis (OA), and even in immune-dysregulating infectious diseases such as the novel coronavirus disease 2019 (COVID-19). This paper discussed the therapeutic applications of MSC secretome and its biomedical aspects related to immune-mediated conditions. Sources for MSC extraction, their migration and homing properties, therapeutic molecules released by MSCs, and the pathways and molecular mechanisms possibly involved in the exceptional immunoregulatory competence of MSCs were discussed. Besides, the novel discoveries and recent findings on immunomodulatory plasticity of MSCs, clinical applications, and the methods required for their use as an effective therapeutic option in patients with immune-mediated/immune-dysregulating diseases were highlighted.
Collapse
Affiliation(s)
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Surendar Aravindhan
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Angelina Olegovna Zekiy
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit (G401), 69120 Heidelberg, Germany
| | | | - Yashwant Pathak
- Professor and Associate Dean for Faculty Affairs, Taneja College of Pharmacy, University of South Florida, Tampa, FL USA
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Shamlou
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Regenerative Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Girousse A, Mathieu M, Sastourné-Arrey Q, Monferran S, Casteilla L, Sengenès C. Endogenous Mobilization of Mesenchymal Stromal Cells: A Pathway for Interorgan Communication? Front Cell Dev Biol 2021; 8:598520. [PMID: 33490065 PMCID: PMC7820193 DOI: 10.3389/fcell.2020.598520] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
To coordinate specialized organs, inter-tissue communication appeared during evolution. Consequently, individual organs communicate their states via a vast interorgan communication network (ICN) made up of peptides, proteins, and metabolites that act between organs to coordinate cellular processes under homeostasis and stress. However, the nature of the interorgan signaling could be even more complex and involve mobilization mechanisms of unconventional cells that are still poorly described. Mesenchymal stem/stromal cells (MSCs) virtually reside in all tissues, though the biggest reservoir discovered so far is adipose tissue where they are named adipose stromal cells (ASCs). MSCs are thought to participate in tissue maintenance and repair since the administration of exogenous MSCs is well known to exert beneficial effects under several pathological conditions. However, the role of endogenous MSCs is barely understood. Though largely debated, the presence of circulating endogenous MSCs has been reported in multiple pathophysiological conditions, but the significance of such cell circulation is not known and therapeutically untapped. In this review, we discuss current knowledge on the circulation of native MSCs, and we highlight recent findings describing MSCs as putative key components of the ICN.
Collapse
Affiliation(s)
- Amandine Girousse
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| | - Maxime Mathieu
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| | - Quentin Sastourné-Arrey
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Sylvie Monferran
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| | - Louis Casteilla
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| | - Coralie Sengenès
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
16
|
Li YL, Chen CH, Chen JY, Lai YS, Wang SC, Jiang SS, Hung WC. Single-cell analysis reveals immune modulation and metabolic switch in tumor-draining lymph nodes. Oncoimmunology 2020; 9:1830513. [PMID: 33117603 PMCID: PMC7575008 DOI: 10.1080/2162402x.2020.1830513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Lymph-node metastasis is a prognosis factor for poor clinical outcome of breast cancer patients. Currently, how breast cancer cells establish pre-metastatic niche in the tumor-draining lymph nodes (TDLNs) is still unclear. To address this question, we isolated heterogeneous cells including immune and stromal cells from naive lymph nodes (LNs) of the FVB/NJ mice and TDLNs of the MMTV-PyMT mice. Single-cell RNA sequencing was performed to investigate the transcriptome of the cells and various bioinformatics analyses were used to identify the altered pathways. Our results revealed several significant changes between naïve LNs and TDLNs. First, according to immunologic signature and pathway analysis, CD4+ and CD8 + T cells showed upregulated angiogenesis pathway genes and higher regulatory T (Treg)-associated genes while they demonstrated downregulation of interferon response and inflammatory response gene signatures, concurrently suggesting an immunosuppressive microenvironment in the TDLNs. Second, profiling of B cells showed down-regulation of marginal zone B lymphocytes in the TDLNs, which was validated by flow cytometric analysis. Third, we found the enhancement of oxidative phosphorylation pathway in the fibroblastic reticular cells (FRCs) of the MMTV-PyMT mice and the elevation of related genes including Prdx3, Ndufa4 and Uqcrb, suggesting massive ATP consumption and TCA cycle metabolism in the FRCs. Collectively, our results reveal the reprogramming of TDLNs during breast cancer progression at single-cell level in a spontaneous breast cancer model and suggest the changes in immune modulation and metabolic switch are key alterations in the preparation of pre-metastatic niche by breast cancer cells.
Collapse
Affiliation(s)
- Yen-Liang Li
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Chung-Hsing Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Jing-Yi Chen
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - You-Syuan Lai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Shao-Chun Wang
- Graduate Institute of Biomedical Sciences, and the Graduate Program of Cancer Biology and Drug Development, China Medical University, Taichung, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Shih-Sheng Jiang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
17
|
The Release of Adipose Stromal Cells from Subcutaneous Adipose Tissue Regulates Ectopic Intramuscular Adipocyte Deposition. Cell Rep 2020; 27:323-333.e5. [PMID: 30970240 DOI: 10.1016/j.celrep.2019.03.038] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 12/30/2018] [Accepted: 03/11/2019] [Indexed: 02/01/2023] Open
Abstract
Ectopic lipid deposition (ELD) is defined by excess fat storage in locations not classically associated with adipose tissue (AT) storage. ELD is positively correlated with insulin resistance and increased risk of metabolic disorders. ELD appears as lipid droplets or adipocytes, whose cell origin is unknown. We previously showed that subcutaneous AT (ScAT) releases adipocyte progenitors into the circulation. Here, we demonstrate that triggering or preventing the release of adipocyte precursors from ScAT directly promoted or limited ectopic adipocyte formation in skeletal muscle in mice. Importantly, obesity-associated metabolic disorders could be mimicked by causing adipocyte precursor release without a high-fat diet. Finally, during nutrient overload, adipocyte progenitors exited ScAT, where their retention signals (CXCR4/CXCL12 axis) were greatly decreased, and further infiltrated skeletal muscles. These data provide insights into the formation of ELD associated with calorie overload and highlight adipocyte progenitor trafficking as a potential target in the treatment of metabolic diseases.
Collapse
|
18
|
di Somma M, Vliora M, Grillo E, Castro B, Dakou E, Schaafsma W, Vanparijs J, Corsini M, Ravelli C, Sakellariou E, Mitola S. Role of VEGFs in metabolic disorders. Angiogenesis 2019; 23:119-130. [PMID: 31853841 DOI: 10.1007/s10456-019-09700-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023]
Abstract
Obesity and metabolic disorders are important public health problems. In this review, the role of vasculature network and VEGF in the adipose tissue maintenance and supplementation is discussed. Angiogenesis is a key process implicated in regulation of tissues homeostasis. Dysregulation of new blood vessels formation may be crucial and contribute to the onset of several pathological conditions, including metabolic syndrome-associated disorders. Adipose tissue homeostasis is fine regulated by vascular network. Vessels support adipose structure. Vasculature modulates the balance between positive and negative regulator factors. In white adipose tissue, vascular endothelial growth factor (VEGF) controls the metabolic activities of adipocytes promoting the trans-differentiation from white to beige phenotype. Trans-differentiation results in an increase of energy consumption. VEGF exerts an opposite effect on brown adipose tissue, where VEGF increases oxygen supply and improves energy expenditure inducing the whitening of adipocytes.
Collapse
Affiliation(s)
- M di Somma
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - M Vliora
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece
| | - E Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - B Castro
- Histocell, S.L.Parque Tecnológico 801A, 2º, 48160, Derio, Bizkaia, Spain
| | - E Dakou
- Laboratory of Cell Genetics, Department of Biology, Faculty of Science and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - W Schaafsma
- Histocell, S.L.Parque Tecnológico 801A, 2º, 48160, Derio, Bizkaia, Spain
| | - J Vanparijs
- Laboratory of Cell Genetics, Department of Biology, Faculty of Science and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - M Corsini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - C Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - E Sakellariou
- FAME Laboratory, Department of Exercise Science, University of Thessaly, Trikala, Greece
| | - S Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
19
|
Ehsanipour A, Nguyen T, Aboufadel T, Sathialingam M, Cox P, Xiao W, Walthers CM, Seidlits SK. Injectable, Hyaluronic Acid-Based Scaffolds with Macroporous Architecture for Gene Delivery. Cell Mol Bioeng 2019; 12:399-413. [PMID: 31719923 PMCID: PMC6816628 DOI: 10.1007/s12195-019-00593-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/20/2019] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Biomaterials can provide localized reservoirs for controlled release of therapeutic biomolecules and drugs for applications in tissue engineering and regenerative medicine. As carriers of gene-based therapies, biomaterial scaffolds can improve efficiency and delivery-site localization of transgene expression. Controlled delivery of gene therapy vectors from scaffolds requires cell-scale macropores to facilitate rapid host cell infiltration. Recently, advanced methods have been developed to form injectable scaffolds containing cell-scale macropores. However, relative efficacy of in vivo gene delivery from scaffolds formulated using these general approaches has not been previously investigated. Using two of these methods, we fabricated scaffolds based on hyaluronic acid (HA) and compared how their unique, macroporous architectures affected their respective abilities to deliver transgenes via lentiviral vectors in vivo. METHODS Three types of scaffolds-nanoporous HA hydrogels (NP-HA), annealed HA microparticles (HA-MP) and nanoporous HA hydrogels containing protease-degradable poly(ethylene glycol) (PEG) microparticles as sacrificial porogens (PEG-MP)-were loaded with lentiviral particles encoding reporter transgenes and injected into mouse mammary fat. Scaffolds were evaluated for their ability to induce rapid infiltration of host cells and subsequent transgene expression. RESULTS Cell densities in scaffolds, distances into which cells penetrated scaffolds, and transgene expression levels significantly increased with delivery from HA-MP, compared to NP-HA and PEG-MP, scaffolds. Nearly 8-fold greater cell densities and up to 16-fold greater transgene expression levels were found in HA-MP, over NP-HA, scaffolds. Cell profiling revealed that within HA-MP scaffolds, macrophages (F4/80+), fibroblasts (ERTR7+) and endothelial cells (CD31+) were each present and expressed delivered transgene. CONCLUSIONS Results demonstrate that injectable scaffolds containing cell-scale macropores in an open, interconnected architecture support rapid host cell infiltration to improve efficiency of biomaterial-mediated gene delivery.
Collapse
Affiliation(s)
- Arshia Ehsanipour
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Tommy Nguyen
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Tasha Aboufadel
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Mayilone Sathialingam
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Phillip Cox
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Weikun Xiao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Christopher M. Walthers
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
| | - Stephanie K. Seidlits
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095 USA
- Broad Stem Cell Research Center, University of California Los Angeles, Los Angeles, CA 90095 USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095 USA
- Brain Research Institute, University of California Los Angeles, Los Angeles, CA 90095 USA
- Center for Minimally Invasive Therapeutics, University of California Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
20
|
Rabhi N, Farmer SR. Adipose Progenitor Cells Contribute to Lipid Spillover during Obesity. Trends Endocrinol Metab 2019; 30:416-418. [PMID: 31153731 DOI: 10.1016/j.tem.2019.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 05/14/2019] [Indexed: 01/16/2023]
Abstract
A recent study (Girousse et al. Cell Rep. 2019;27:323-333) shows that CXCR4+ adipose progenitors (APCs) contribute to lipid spillover during high-fat feeding through their release from subcutaneous fat depots (ScATs) and migration to skeletal muscle where they differentiate into adipocytes. Pharmacological antagonism of CXCR4, which prevents the CXCL12-dependent retention of APCs in ScAT, mimics the effects of overfeeding.
Collapse
Affiliation(s)
- Nabil Rabhi
- Department of Biochemistry, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Stephen R Farmer
- Department of Biochemistry, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA.
| |
Collapse
|
21
|
Plaza A, Merino B, Del Olmo N, Ruiz-Gayo M. The cholecystokinin receptor agonist, CCK-8, induces adiponectin production in rat white adipose tissue. Br J Pharmacol 2019; 176:2678-2690. [PMID: 31012948 DOI: 10.1111/bph.14690] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE A cholecystokinin (CCK) system has been identified in white adipose tissue (WAT). Nevertheless, the endocrine actions of CCK on WAT remain unknown. Our goal was to investigate the role of CCK in regulating the production of adiponectin, an adipokine expressed in WAT, which is pivotal in preserving energy homeostasis. EXPERIMENTAL APPROACH The effect of the bioactive CCK fragment CCK-8 on adiponectin production was studied both in vivo and in vitro. CCK-8 effects were characterized in rats treated with selective CCK1 and CCK2 receptor antagonists as well as in pre-adipocytes carrying the selective silencing of either CCK1 or CCK2 receptors. The influence of insulin on CCK-8 responses was also analysed. KEY RESULTS In WAT, CCK-8 increased plasma adiponectin levels and the expression of the adiponectin gene (Adipoq). In pre-adipocytes, CCK-8 up-regulated adiponectin production. CCK-8 effects were abolished by L-365,260, a selective CCK2 receptor antagonist. CCK2 receptor knockdown also abolished the effects of CCK-8 in pre-adipocytes. Moreover, in vitro CCK-8 effects were blocked by triciribine, a specific inhibitor of protein kinase B (Akt) and by the PPARγ antagonist T0070907. Silencing the expression of the insulin receptor inhibited CCK-8-induced Adipoq expression in pre-adipocytes. Furthermore, insulin potentiated the effect of CCK-8. CONCLUSION AND IMPLICATIONS CCK-8 stimulates adiponectin production in WAT by acting on CCK2 receptors, through a mechanism involving both Akt and PPARγ. Moreover, CCK-8 actions are only observed in the presence of insulin. Our results could have translational value in the design of new insulin-sensitizing therapies.
Collapse
Affiliation(s)
- Adrián Plaza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
| | - Beatriz Merino
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
| | - Nuria Del Olmo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
| | - Mariano Ruiz-Gayo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU San Pablo, Madrid, Spain
| |
Collapse
|
22
|
Making Sense of Stem Cells and Fat Grafting in Plastic Surgery: The Hype, Evidence, and Evolving U.S. Food and Drug Administration Regulations. Plast Reconstr Surg 2019; 143:417e-424e. [PMID: 30688913 DOI: 10.1097/prs.0000000000005207] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Autologous fat grafting and adipose-derived stem cells are two distinct entities with two different risk profiles, and should be regulated as such. Autologous fat grafting prepared with the additional step of stromal vascular fraction isolation is considered a form of "stem cell therapy" given the high concentration of stem cells found in stromal vascular fraction. Much ambiguity existed in the distinction between autologous fat grafting and stromal vascular fraction initially, in terms of both their biological properties and how they should be regulated. The market has capitalized on this in the past decade to sell unproven "stem cell" therapies to unknowing consumers while exploiting the regulatory liberties of traditional fat grafting. This led to a Draft Guidance from the U.S. Food and Drug Administration in 2014 proposing stricter regulations on fat grafting in general, which in turn elicited a response from plastic surgeons, who have safely used autologous fat grafting in the clinical setting for over a century. After a series of discussions, the U.S. Food and Drug Administration released its Final Guidance in November of 2017, which established clear distinctions between autologous fat grafting and stromal vascular fraction and their separate regulations. By educating ourselves on the U.S. Food and Drug Administration's final stance on fat grafting and stem cell therapy, we can learn how to navigate the regulatory waters for the two entities and implement their clinical use in a responsible and informed manner.
Collapse
|
23
|
Plaza A, Naranjo V, Blonda AM, Cano V, González-Martín C, Gil-Ortega M, Ruiz-Gayo M, Merino B. Inflammatory stress and altered angiogenesis evoked by very high-fat diets in mouse liver. ACTA ACUST UNITED AC 2019; 66:434-442. [PMID: 30833154 DOI: 10.1016/j.endinu.2018.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 12/19/2018] [Accepted: 12/28/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD), a condition that leads to fibrosis, is caused by intake of very high-fat diets (HFDs). However, while the negative impact on the liver of these diets has been an issue of interest, systematic research on the effect of HFDs are lacking. OBJECTIVE To characterize the overall impact of HFDs on both molecular and morphological signs of liver remodeling. METHODS A study was conducted on male C57BL/6J mice to assess the effect of 4- and 8-week HFDs (60% kcal from fat) on (i) liver steatosis and fibrosis, and (ii) expression of factors involved in inflammation and angiogenesis. RESULTS After an 8-week HFD, vascular endothelial growth factor type-2 receptor (VEGF-R2) and fatty acid translocase/trombospondin-1 receptor (CD36) were overexpressed in liver tissue of mice given HFDs. These changes suggest impaired liver angiogenesis and occurred together with (i) increased GPR78-BiP and EIF2α phosphorylation, suggesting endoplasmic reticulum stress, (ii) induction of Col1a1 gene expression, a marker of fibrosis, and (iii) increased CD31 immunolabeling, consistent with active angiogenesis and fibrosis. CONCLUSION Our data show that very HFDs promote a rapid inflammatory response, as well as deregulation of angiogenesis, both consistent with development of liver fibrosis.
Collapse
Affiliation(s)
- Adrián Plaza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| | - Víctor Naranjo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| | - Alessandra M Blonda
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| | - Victoria Cano
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| | - Carmen González-Martín
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain
| | - Mariano Ruiz-Gayo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain.
| | - Beatriz Merino
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, CEU Universities, Madrid, Spain.
| |
Collapse
|
24
|
Co-transplantation of mesenchymal stem cells improves spermatogonial stem cell transplantation efficiency in mice. Stem Cell Res Ther 2018; 9:317. [PMID: 30463610 PMCID: PMC6249754 DOI: 10.1186/s13287-018-1065-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/19/2018] [Accepted: 10/31/2018] [Indexed: 12/27/2022] Open
Abstract
Background Spermatogonial stem cell transplantation (SSCT) could become a fertility restoration tool for childhood cancer survivors. However, since in mice, the colonization efficiency of transplanted spermatogonial stem cells (SSCs) is only 12%, the efficiency of the procedure needs to be improved before clinical implementation is possible. Co-transplantation of mesenchymal stem cells (MSCs) might increase colonization efficiency of SSCs by restoring the SSC niche after gonadotoxic treatment. Methods A mouse model for long-term infertility was developed and used to transplant SSCs (SSCT, n = 10), MSCs (MSCT, n = 10), a combination of SSCs and MSCs (MS-SSCT, n = 10), or a combination of SSCs and TGFß1-treated MSCs (MSi-SSCT, n = 10). Results The best model for transplantation was obtained after intraperitoneal injection of busulfan (40 mg/kg body weight) at 4 weeks followed by CdCl2 (2 mg/kg body weight) at 8 weeks of age and transplantation at 11 weeks of age. Three months after transplantation, spermatogenesis resumed with a significantly better tubular fertility index (TFI) in all transplanted groups compared to non-transplanted controls (P < 0.001). TFI after MSi-SSCT (83.3 ± 19.5%) was significantly higher compared to MS-SSCT (71.5 ± 21.7%, P = 0.036) but did not differ statistically compared to SSCT (78.2 ± 12.5%). In contrast, TFI after MSCT (50.2 ± 22.5%) was significantly lower compared to SSCT (P < 0.001). Interestingly, donor-derived TFI was found to be significantly improved after MSi-SSCT (18.8 ± 8.0%) compared to SSCT (1.9 ± 1.1%; P < 0.001), MSCT (0.0 ± 0.0%; P < 0.001), and MS-SSCT (3.4 ± 1.9%; P < 0.001). While analyses showed that both native and TGFß1-treated MSCs maintained characteristics of MSCs, the latter showed less migratory characteristics and was not detected in other organs. Conclusion Co-transplanting SSCs and TGFß1-treated MSCs significantly improves the recovery of endogenous SSCs and increases the homing efficiency of transplanted SSCs. This procedure could become an efficient method to treat infertility in a clinical setup, once the safety of the technique has been proven. Electronic supplementary material The online version of this article (10.1186/s13287-018-1065-0) contains supplementary material, which is available to authorized users.
Collapse
|
25
|
Expression analysis of a cholecystokinin system in human and rat white adipose tissue. Life Sci 2018; 206:98-105. [PMID: 29800537 DOI: 10.1016/j.lfs.2018.05.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/13/2018] [Accepted: 05/21/2018] [Indexed: 01/22/2023]
Abstract
AIM Cholecystokinin (CCK) participates in the storage of dietary triglycerides in white adipose tissue (WAT). Our goal was to characterize, both in subcutaneous (Sc-WAT) and visceral WAT (Vis-WAT), the functional expression of the two known CCK receptors, CCK-1 (CCK-1R) and CCK-2 (CCK-2R), as well as of CCK. MAIN METHODS Gene and protein expression was assessed in different cell types of rat and human WAT by means of RT-PCR and western-blot, respectively. The functionality of CCK-Rs was tested by quantifying protein kinase B (Akt) phosphorylation after treatment of pre-adipocytes with the bioactive fragment of CCK, CCK-8. The CCK receptor subtype involved in Akt phosphorylation was investigated by using selective CCK-1R (SR-27,897) and CCK-2R antagonists (L-365,260). KEY FINDINGS In rats, CCK-1R (Cckar) and CCK-2R (Cckbr) gene expression was detected in the two types of WAT analyzed as well as in isolated adipocytes, mesenchymal stem cells and pre-adipocytes. CCK-1R and CCK-2R proteins were identified in adipocytes and, to a minor extent, in pre-adipocytes. In addition, CCK-2R were detected in subcutaneous mesenchymal stem cells. Gene expression of the CCK precursor preproCCK as well as CCK immunoreactivity were also found in Sc-WAT and Vis-WAT. In human WAT, CCK gene expression as well as CCK-2Rs and CCK were also identified. CCK-8 evoked Akt phosphorylation in rat pre-adipocytes, and this effect was antagonized by SR-27,897 and L-365,260. SIGNIFICANCE Our data show that both human and rat WAT express a complete CCK system, and suggest that CCK may have an autocrine/paracrine role in regulating adipose tissue biology.
Collapse
|
26
|
de Lucas B, Pérez LM, Gálvez BG. Importance and regulation of adult stem cell migration. J Cell Mol Med 2017; 22:746-754. [PMID: 29214727 PMCID: PMC5783855 DOI: 10.1111/jcmm.13422] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/14/2017] [Indexed: 12/13/2022] Open
Abstract
Cell migration is an essential process throughout the life of vertebrates, beginning during embryonic development and continuing throughout adulthood. Stem cells have an inherent ability to migrate, that is as important as their capacity for self‐renewal and differentiation, enabling them to maintain tissue homoeostasis and mediate repair and regeneration. Adult stem cells reside in specific tissue niches, where they remain in a quiescent state until called upon and activated by tissue environmental signals. Cell migration is a highly regulated process that involves the integration of intrinsic signals from the niche and extrinsic factors. Studies using three‐dimensional in vitro models have revealed the astonishing plasticity of cells in terms of the migration modes employed in response to changes in the microenvironment. These same properties can, however, be subverted during the development of some pathologies such as cancer. In this review, we describe the response of adult stem cells to migratory stimuli and the mechanisms by which they sense and transduce intracellular signals involved in migratory processes. Understanding the molecular events underlying migration may help develop therapeutic strategies for regenerative medicine and to treat diseases with a cell migration component.
Collapse
Affiliation(s)
- Beatriz de Lucas
- Universidad Europea de Madrid, Madrid, Spain.,Instituto de Investigación Hospital, 12 de Octubre, Madrid, Spain
| | - Laura M Pérez
- Universidad Europea de Madrid, Madrid, Spain.,Instituto de Investigación Hospital, 12 de Octubre, Madrid, Spain
| | - Beatriz G Gálvez
- Universidad Europea de Madrid, Madrid, Spain.,Instituto de Investigación Hospital, 12 de Octubre, Madrid, Spain
| |
Collapse
|
27
|
Gaborit B, Sengenes C, Ancel P, Jacquier A, Dutour A. Role of Epicardial Adipose Tissue in Health and Disease: A Matter of Fat? Compr Physiol 2017. [PMID: 28640452 DOI: 10.1002/cphy.c160034] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Epicardial adipose tissue (EAT) is a small but very biologically active ectopic fat depot that surrounds the heart. Given its rapid metabolism, thermogenic capacity, unique transcriptome, secretory profile, and simply measurability, epicardial fat has drawn increasing attention among researchers attempting to elucidate its putative role in health and cardiovascular diseases. The cellular crosstalk between epicardial adipocytes and cells of the vascular wall or myocytes is high and suggests a local role for this tissue. The balance between protective and proinflammatory/profibrotic cytokines, chemokines, and adipokines released by EAT seem to be a key element in atherogenesis and could represent a future therapeutic target. EAT amount has been found to predict clinical coronary outcomes. EAT can also modulate cardiac structure and function. Its amount has been associated with atrial fibrillation, coronary artery disease, and sleep apnea syndrome. Conversely, a beiging fat profile of EAT has been identified. In this review, we describe the current state of knowledge regarding the anatomy, physiology and pathophysiological role of EAT, and the factors more globally leading to ectopic fat development. We will also highlight the most recent findings on the origin of this ectopic tissue, and its association with cardiac diseases. © 2017 American Physiological Society. Compr Physiol 7:1051-1082, 2017.
Collapse
Affiliation(s)
- Bénédicte Gaborit
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,Endocrinology Metabolic Diseases, and Nutrition Department, Pole ENDO, APHM, Aix-Marseille Univ, Marseille, France
| | - Coralie Sengenes
- STROMALab, Université de Toulouse, EFS, ENVT, Inserm U1031, ERL CNRS 5311, CHU Rangueil, Toulouse, France
| | - Patricia Ancel
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France
| | - Alexis Jacquier
- CNRS UMR 7339, Centre de Résonance Magnétique Biologique et Médicale (CRMBM), Marseille, France.,Radiology department, CHU La Timone, Marseille, France
| | - Anne Dutour
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,Endocrinology Metabolic Diseases, and Nutrition Department, Pole ENDO, APHM, Aix-Marseille Univ, Marseille, France
| |
Collapse
|
28
|
Lopez-Santalla M, Mancheño-Corvo P, Escolano A, Menta R, DelaRosa O, Abad JL, Büscher D, Redondo JM, Bueren JA, Dalemans W, Lombardo E, Garin MI. Biodistribution and Efficacy of Human Adipose-Derived Mesenchymal Stem Cells Following Intranodal Administration in Experimental Colitis. Front Immunol 2017. [PMID: 28642759 PMCID: PMC5462906 DOI: 10.3389/fimmu.2017.00638] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have a large potential in cell therapy for treatment of inflammatory and autoimmune diseases, thanks to their immunomodulatory properties. The encouraging results in animal models have initiated the translation of MSC therapy to clinical trials. In cell therapy protocols with MSCs, administered intravenously, several studies have shown that a small proportion of infused MSCs can traffic to the draining lymph nodes (LNs). This is accompanied with an increase of different types of regulatory immune cells in the LNs, suggesting the importance of migration of MSCs to the LNs in order to contribute to immunomodulatory response. Intranodal (IN), also referred as intralymphatic, injection of cells, like dendritic cells, is being proposed in the clinic for the treatment of cancer and allergy, showing that this route of administration is clinically safe and efficient. In this study, we investigated, for the first time, the biodistribution and the efficacy of Luciferase+ adipose-derived MSCs (Luci-eASCs), infused through the inguinal LNs (iLNs), in normal mice and in inflamed mice with colitis. Most of the Luci-eASCs remain in the iLNs and in the adipose tissue surrounding the inguinal LNs. A small proportion of Luci-eASCs can migrate to other locations within the lymphatic system and to other tissues and organs, having a preferential migration toward the intestine in colitic mice. Our results show that the infused Luci-eASCs protected 58% of the mice against induced colitis. Importantly, a correlation between the response to eASC treatment and a higher accumulation of eASCs in popliteal, parathymic, parathyroid, and mesenteric LNs were found. Altogether, these results suggest that IN administration of eASCs is feasible and may represent an effective strategy for cell therapy protocols with human adipose-derived MSCs in the clinic for the treatment of immune-mediated disorders.
Collapse
Affiliation(s)
- Mercedes Lopez-Santalla
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | | - Amelia Escolano
- Gene Regulation in Cardiovascular Remodeling and Inflammation Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | | | | | | | | | - Juan M Redondo
- Gene Regulation in Cardiovascular Remodeling and Inflammation Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Juan A Bueren
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | | | | - Marina I Garin
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| |
Collapse
|
29
|
Salazar TE, Richardson MR, Beli E, Ripsch MS, George J, Kim Y, Duan Y, Moldovan L, Yan Y, Bhatwadekar A, Jadhav V, Smith JA, McGorray S, Bertone AL, Traktuev DO, March KL, Colon-Perez LM, Avin K, Sims E, Mund JA, Case J, Deng S, Kim MS, McDavitt B, Boulton ME, Thinschmidt J, Calzi SL, Fitz SD, Fuchs RK, Warden SJ, McKinley T, Shekhar A, Febo M, Johnson PL, Chang LJ, Gao Z, Kolonin MG, Lai S, Ma J, Dong X, White FA, Xie H, Yoder MC, Grant MB. Electroacupuncture Promotes Central Nervous System-Dependent Release of Mesenchymal Stem Cells. Stem Cells 2017; 35:1303-1315. [PMID: 28299842 PMCID: PMC5530374 DOI: 10.1002/stem.2613] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/16/2017] [Accepted: 02/11/2017] [Indexed: 12/12/2022]
Abstract
Electroacupuncture (EA) performed in rats and humans using limb acupuncture sites, LI-4 and LI-11, and GV-14 and GV-20 (humans) and Bai-hui (rats) increased functional connectivity between the anterior hypothalamus and the amygdala and mobilized mesenchymal stem cells (MSCs) into the systemic circulation. In human subjects, the source of the MSC was found to be primarily adipose tissue, whereas in rodents the tissue sources were considered more heterogeneous. Pharmacological disinhibition of rat hypothalamus enhanced sympathetic nervous system (SNS) activation and similarly resulted in a release of MSC into the circulation. EA-mediated SNS activation was further supported by browning of white adipose tissue in rats. EA treatment of rats undergoing partial rupture of the Achilles tendon resulted in reduced mechanical hyperalgesia, increased serum interleukin-10 levels and tendon remodeling, effects blocked in propranolol-treated rodents. To distinguish the afferent role of the peripheral nervous system, phosphoinositide-interacting regulator of transient receptor potential channels (Pirt)-GCaMP3 (genetically encoded calcium sensor) mice were treated with EA acupuncture points, ST-36 and LIV-3, and GV-14 and Bai-hui and resulted in a rapid activation of primary sensory neurons. EA activated sensory ganglia and SNS centers to mediate the release of MSC that can enhance tissue repair, increase anti-inflammatory cytokine production and provide pronounced analgesic relief. Stem Cells 2017;35:1303-1315.
Collapse
Affiliation(s)
- Tatiana E. Salazar
- Genetics Institute, University of Florida, Gainesville, FL 32610, USA
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Matthew R. Richardson
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Eleni Beli
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Matthew S. Ripsch
- Department of Anesthesia, Indiana University, Indianapolis, IN 46202, USA
| | - John George
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Youngsook Kim
- Department of Anesthesia, Indiana University, Indianapolis, IN 46202, USA
| | - Yaqian Duan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Leni Moldovan
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yuanqing Yan
- Genetics Institute, University of Florida, Gainesville, FL 32610, USA
| | - Ashay Bhatwadekar
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Vaishnavi Jadhav
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jared A. Smith
- Department of Anesthesia, Indiana University, Indianapolis, IN 46202, USA
| | - Susan McGorray
- Department of Biostatistics, University of Florida, Gainesville, FL 32610, USA
| | - Alicia L. Bertone
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Dmitri O. Traktuev
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN 46202, USA
- Indiana Center for Vascular Biology and Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Keith L. March
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN 46202, USA
- Indiana Center for Vascular Biology and Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Luis M. Colon-Perez
- Department of Psychiatry, University of Florida, McKnight Brain Institute, Gainesville, FL 32610, USA
| | - Keith Avin
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN 46202, USA
| | - Emily Sims
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Julie A. Mund
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Jamie Case
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA
- Scripps Clinic Medical Group, Scripps Center for Organ and Cell Transplantation, La Jolla, CA 92037, USA
- Department of Pediatrics, Indiana University, Indianapolis, IN 46202USA
| | - Shaolin Deng
- Mainland Acupuncture, Gainesville, FL 32653, USA
| | - Min Su Kim
- College of Veterinary Medicine, Chon Buk National University, Jeonju, South Korea
| | | | - Michael E. Boulton
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jeffrey Thinschmidt
- Department of Pharmacology, University of Florida, Gainesville, FL 32610, USA
| | - Sergio Li Calzi
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Stephanie D. Fitz
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN 46202, USA
| | - Robyn K. Fuchs
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN 46202, USA
| | - Stuart J. Warden
- Department of Physical Therapy, Indiana University School of Health and Rehabilitation Sciences, Indianapolis, IN 46202, USA
| | - Todd McKinley
- Department of Orthopedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Anantha Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, McKnight Brain Institute, Gainesville, FL 32610, USA
| | - Phillip L. Johnson
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lung Ji Chang
- Department of Molecular Genetics & Microbiology, University of Florida, Gainesville, FL 32611, USA
| | - Zhanguo Gao
- Center for Metabolic and Degenerative Diseases, Harry E. Bovay Institute of Molecular Medicine University of Texas Health Science Center, Houston, TX 77030, USA
| | - Mikhail G. Kolonin
- Center for Metabolic and Degenerative Diseases, Harry E. Bovay Institute of Molecular Medicine University of Texas Health Science Center, Houston, TX 77030, USA
| | - Song Lai
- Department of Radiation Oncology, University of Florida School of Medicine, Gainesville, FL 32610, USA
| | - Jinfeng Ma
- Department of Radiation Oncology, University of Florida School of Medicine, Gainesville, FL 32610, USA
| | - Xinzhong Dong
- Department of Neuroscience, Center of Sensory Biology, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Fletcher A. White
- Department of Anesthesia, Indiana University, Indianapolis, IN 46202, USA
| | - Huisheng Xie
- College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| | - Mervin C. Yoder
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Maria B. Grant
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
30
|
Mancheño-Corvo P, Lopez-Santalla M, Menta R, DelaRosa O, Mulero F, Del Rio B, Ramirez C, Büscher D, Bueren JA, Lopez-Belmonte J, Dalemans W, Garin MI, Lombardo E. Intralymphatic Administration of Adipose Mesenchymal Stem Cells Reduces the Severity of Collagen-Induced Experimental Arthritis. Front Immunol 2017; 8:462. [PMID: 28484460 PMCID: PMC5399019 DOI: 10.3389/fimmu.2017.00462] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 04/04/2017] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells with immunomodulatory properties. They have emerged as a very promising treatment for autoimmunity and inflammatory diseases such as rheumatoid arthritis. Previous studies have demonstrated that MSCs, administered systemically, migrate to lymphoid tissues associated with the inflammatory site where functional MSC-induced immune cells with a regulatory phenotype were increased mediating the immunomodulatory effects of MSCs. These results suggest that homing of MSCs to the lymphatic system plays an important role in the mechanism of action of MSCs in vivo. Thus, we hypothesized that direct intralymphatic (IL) (also referred as intranodal) administration of MSCs could be an alternative and effective route of administration for MSC-based therapy. Here, we report the feasibility and efficacy of the IL administration of human expanded adipose mesenchymal stem cells (eASCs) in a mouse model of collagen-induced arthritis (CIA). IL administration of eASCs attenuated the severity and progression of arthritis, reduced bone destruction and increased the levels of regulatory T cells (CD25+Foxp3+CD4+ cells) and Tr1 cells (IL10+CD4+), in spleen and draining lymph nodes. Taken together, these results indicate that IL administration of eASCs is very effective in modulating established CIA and may represent an alternative treatment modality for cell therapy with eASCs.
Collapse
Affiliation(s)
| | - Mercedes Lopez-Santalla
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | | | | - Francisca Mulero
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | | | | | | | - Juan A Bueren
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | | | | - Marina I Garin
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | |
Collapse
|
31
|
Valencia J, Jiménez E, Martínez VG, Del Amo BG, Hidalgo L, Entrena A, Fernández-Sevilla LM, Del Río F, Varas A, Vicente Á, Sacedón R. Characterization of human fibroblastic reticular cells as potential immunotherapeutic tools. Cytotherapy 2017; 19:640-653. [PMID: 28262465 DOI: 10.1016/j.jcyt.2017.01.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/26/2017] [Accepted: 01/31/2017] [Indexed: 01/09/2023]
Abstract
Fibroblastic reticular cells (FRCs) are essential players during adaptive immune responses not only as a structural support for the encounter of antigen-presenting cells and naive T lymphocytes but also as a source of modulatory signals. However, little is known about this cell population in humans. To address the phenotypical and functional analysis of human FRCs here we established splenic (SP) and mesenteric lymph node (LN) CD45-CD31-CD90+podoplanin+ myofibroblastic cell cultures. They shared the phenotypical characteristics distinctive of FRCs, including the expression of immunomodulatory factors and peripheral tissue antigens. Nevertheless, human FRCs also showed particular features, some differing from mouse FRCs, like the lack of nitric oxide synthase (NOS2) expression after interferon (IFN)γstimulation. Interestingly, SP-FRCs expressed higher levels of interleukin (IL)-6, BMP4, CCL2, CXCL12 and Notch molecules, and strongly adapted their functional profile to lipopolysaccharide (LPS), polyinosinic:polycytidylic acid (Poly I:C) and IFNγ stimulation. In contrast, we found higher expression of transforming growth factor (TGF)β and Activin A in LN-FRCs that barely responded via Toll-Like Receptor (TLR)3 and constitutively expressed retinaldehyde dehydrogenase 1 enzyme, absent in SP-FRCs. This study reveals human FRCs can be valuable models to increase our knowledge about the physiology of human secondary lymphoid organs in health and disease and to explore the therapeutic options of FRCs.
Collapse
Affiliation(s)
- Jaris Valencia
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Eva Jiménez
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Víctor G Martínez
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Beatriz G Del Amo
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Laura Hidalgo
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Ana Entrena
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | | | - Francisco Del Río
- Department of Medicine, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Alberto Varas
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Ángeles Vicente
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain
| | - Rosa Sacedón
- Department of Cell Biology, Faculty of Medicine, Complutense University, Madrid, Spain.
| |
Collapse
|
32
|
Lemaitre M, Monsarrat P, Blasco‐Baque V, Loubières P, Burcelin R, Casteilla L, Planat‐Bénard V, Kémoun P. Periodontal Tissue Regeneration Using Syngeneic Adipose-Derived Stromal Cells in a Mouse Model. Stem Cells Transl Med 2016; 6:656-665. [PMID: 28191762 PMCID: PMC5442818 DOI: 10.5966/sctm.2016-0028] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 08/03/2016] [Indexed: 12/18/2022] Open
Abstract
Current treatment of periodontitis is still associated with a high degree of variability in clinical outcomes. Recent advances in regenerative medicine by mesenchymal cells, including adipose stromal cells (ASC) have paved the way to improved periodontal regeneration (PD) but little is known about the biological processes involved. Here, we aimed to use syngeneic ASCs for periodontal regeneration in a new, relevant, bacteria‐induced periodontitis model in mice. Periodontal defects were induced in female C57BL6/J mice by oral gavage with periodontal pathogens. We grafted 2 × 105 syngeneic mouse ASCs expressing green fluorescent protein (GFP) (GFP+/ASC) within a collagen vehicle in the lingual part of the first lower molar periodontium (experimental) while carrier alone was implanted in the contralateral side (control). Animals were sacrificed 0, 1, 6, and 12 weeks after treatment by GFP+/ASC or vehicle graft, and microscopic examination, immunofluorescence, and innovative bio‐informatics histomorphometry methods were used to reveal deep periodontium changes. From 1 to 6 weeks after surgery, GFP+ cells were identified in the periodontal ligament (PDL), in experimental sites only. After 12 weeks, cementum regeneration, the organization of PDL fibers, the number of PD vessels, and bone morphogenetic protein‐2 and osteopontin expression were greater in experimental sites than in controls. Specific stromal cell subsets were recruited in the newly formed tissue in ASC‐implanted periodontium only. These data suggest that ASC grafting in diseased deep periodontium, relevant to human pathology, induces a significant improvement of the PDL microenvironment, leading to a recovery of tooth‐supporting tissue homeostasis. Stem Cells Translational Medicine2017;6:656–665
Collapse
Affiliation(s)
- Mathieu Lemaitre
- Department of Biological Sciences, Dental Faculty, Toulouse University Hospital, University of Toulouse, Toulouse, France
- CNRS ERL 5311, EFS, INPENVT, INSERM U1031, UPS, STROMALab, University of Toulouse, Toulouse, France
| | - Paul Monsarrat
- CNRS ERL 5311, EFS, INPENVT, INSERM U1031, UPS, STROMALab, University of Toulouse, Toulouse, France
- Department of Anatomical Sciences and Radiology, Dental Faculty, Toulouse University Hospital, University of Toulouse, Toulouse, France
| | - Vincent Blasco‐Baque
- Department of Biological Sciences, Dental Faculty, Toulouse University Hospital, University of Toulouse, Toulouse, France
- UMR1048, I2MC, UPS, INSERM, University of Toulouse, Toulouse, France
| | - Pascale Loubières
- Department of Biological Sciences, Dental Faculty, Toulouse University Hospital, University of Toulouse, Toulouse, France
- UMR1048, I2MC, UPS, INSERM, University of Toulouse, Toulouse, France
| | - Rémy Burcelin
- UMR1048, I2MC, UPS, INSERM, University of Toulouse, Toulouse, France
| | - Louis Casteilla
- CNRS ERL 5311, EFS, INPENVT, INSERM U1031, UPS, STROMALab, University of Toulouse, Toulouse, France
| | - Valérie Planat‐Bénard
- CNRS ERL 5311, EFS, INPENVT, INSERM U1031, UPS, STROMALab, University of Toulouse, Toulouse, France
| | - Philippe Kémoun
- Department of Biological Sciences, Dental Faculty, Toulouse University Hospital, University of Toulouse, Toulouse, France
| |
Collapse
|
33
|
Abstract
A critical hallmark of adaptive immune responses is the rapid and extensive expansion of lymph nodes. During this process, the complex internal structure of the organs is maintained revealing the existence of mechanisms able to balance lymph node integrity with structural flexibility. This article reviews the extensive architectural remodeling that occurs within lymph nodes during adaptive immune responses and how it is regulated by dendritic cells (DCs). In particular we focus on previously unappreciated functions of DCs in coordinating remodeling of lymph node vasculature, expansion of the fibroblastic reticular network and maintenance of lymphoid stromal phenotypes. Our increased understanding of these processes indicates that DCs need to be viewed not only as key antigen-presenting cells for lymphocytes but also as broad-acting immune sentinels that convey signals to lymphoid organ stroma and thereby facilitate immune response initiation at multiple levels.
Collapse
Affiliation(s)
- Sophie E Acton
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK
- MRC Laboratory of Molecular and Cellular Biology, University College London, London, UK
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK
| |
Collapse
|
34
|
Lee CY, Kang JY, Lim S, Ham O, Chang W, Jang DH. Hypoxic conditioned medium from mesenchymal stem cells promotes lymphangiogenesis by regulation of mitochondrial-related proteins. Stem Cell Res Ther 2016; 7:38. [PMID: 26968383 PMCID: PMC4788827 DOI: 10.1186/s13287-016-0296-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 02/15/2016] [Accepted: 02/22/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Recently, cell-based therapeutic lymphangiogenesis has emerged and provided hope for lymphatic regeneration. Previous studies have demonstrated that secretomes of mesenchymal stem cells (MSCs) facilitate the regeneration of various damaged tissues. This study was conducted to evaluate the lymphangiogenic potential of hypoxic conditioned media (HCM) from MSCs. METHODS To investigate the effects of MSC-secreted factors in starved human lymphatic endothelial cells (hLEC), hLECs were treated with endothelial basal medium (EBM)-2 (control), normoxic conditioned media (NCM), or HCM in vitro and in vivo. RESULTS MSCs expressed lymphangiogenic factors including EGF, FGF2, HGF, IGF-1, and VEGF-A and -C. hLECs were treated with each medium. hLEC proliferation, migration, and tube formation were improved under HCM compared with NCM. Moreover, expression of mitochondrial-related factors, MFN1and 2, were improved in HCM-treated hLECs. Lymphedema mice injected with HCM showed markedly decreased lymphedema via increased lymphatic vessel formation when compared with EBM-2- or NCM-treated mice. CONCLUSIONS This study suggested that HCM from MSCs contain high levels of secreted lymphangiogenic factors and promote lymphangiogenesis by regulating mitochondrial-related factors. Thus, treatment with HCM may be a therapeutic strategy for lymphedema.
Collapse
Affiliation(s)
- Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Graduate School, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jin Young Kang
- Department of Rehabilitation Medicine, National Traffic Injury Rehabilitation Hospital, College of Medicine, The Catholic University of Korea, Yangpyeong-gun, 12564, Republic of Korea
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, 25601, Gangwon-do, Republic of Korea
| | - Onju Ham
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City, 22711, Republic of Korea
| | - Woochul Chang
- Department of Biology Education, College of Education, Pusan National University, Busan, 46241, Republic of Korea.
| | - Dae-Hyun Jang
- Department of Rehabilitation Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Dongsu-ro 56, Bupyeong-gu, Incheon, 21431, Republic of Korea.
| |
Collapse
|
35
|
de Witte SFH, Franquesa M, Baan CC, Hoogduijn MJ. Toward Development of iMesenchymal Stem Cells for Immunomodulatory Therapy. Front Immunol 2016; 6:648. [PMID: 26779185 PMCID: PMC4701910 DOI: 10.3389/fimmu.2015.00648] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/14/2015] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSC) are under development as an immunomodulatory therapy. The anticipated immunomodulatory effects of MSC are broad, from direct inhibition of lymphocyte proliferation, induction of regulatory T and B cells, to resetting the immune system via a hit-and-run principle. There are endless flavors of MSC. Differences between MSC are originating from donors variation, differences in tissue of origin, the effects of culture conditions, and expansion time. Even standard culture conditions change the properties of MSC dramatically and generate MSC that only remotely resemble their in vivo counterparts. Adjustments in culture protocols can further emphasize properties of interest in MSC, thereby generating cells fitted for specific purposes. Culture improved immunomodulatory MSC can be designed to target particular immune disorders. In this review, we describe the observed and the desired immunomodulatory effects of MSC and propose approaches how MSC with optimal immunomodulatory properties can be developed.
Collapse
Affiliation(s)
- Samantha F H de Witte
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center , Rotterdam , Netherlands
| | - Marcella Franquesa
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center , Rotterdam , Netherlands
| | - Carla C Baan
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center , Rotterdam , Netherlands
| | - Martin J Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center , Rotterdam , Netherlands
| |
Collapse
|
36
|
Fadini GP, Bonora BM, Marcuzzo G, Marescotti MC, Cappellari R, Pantano G, Sanzari MC, Duran X, Vendrell J, Plebani M, Avogaro A. Circulating Stem Cells Associate With Adiposity and Future Metabolic Deterioration in Healthy Subjects. J Clin Endocrinol Metab 2015; 100:4570-8. [PMID: 26469382 DOI: 10.1210/jc.2015-2867] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CONTEXT Obesity and metabolic syndrome are associated with mild leukocytosis, but whether hematopoietic stem/progenitor cells (HSPCs) play a role in metabolic deterioration is unknown. OBJECTIVE Our objective was to analyze the cross-sectional and longitudinal associations between CD34(+) HSPCs, adiposity, and metabolic syndrome features. DESIGN This is a cross-sectional study on 242 participants, 155 of whom were followed and included in a longitudinal assessment. SETTING This study took place in a tertiary referral center for metabolic diseases. PARTICIPANTS Healthy working individuals attending a cardiovascular screening program (total n = 3158) and having a baseline measure of circulating CD34(+) cells participated. MAIN OUTCOME MEASURES We collected demographic and anthropometric data, cardiovascular risk factors, and metabolic syndrome parameters. RESULTS Participants (34.7% males, mean age 45.9 ± 0.5 years) were free from diabetes and cardiovascular disease. Cross-sectionally, absolute CD34(+) cell counts were directly correlated with body mass index and waist circumference, inversely correlated with high-density lipoprotein cholesterol and the quantitative insulin sensitivity check index, and were higher in individuals with the metabolic syndrome. The hematopoietic component contributed most to the association of CD34(+) cells with adiposity. During a 6.3-year follow-up, high absolute levels of CD34(+) cells were associated with increasing waist circumference, declining quantitative insulin sensitivity check index and high-density lipoprotein cholesterol, and with incidence of metabolic syndrome. Relative CD34(+) cell counts showed weaker associations with metabolic parameters than absolute levels, but were longitudinally associated with increasing waist circumference and metabolic syndrome development. CONCLUSIONS A mild elevation of circulating CD34(+) progenitor cells, reflecting expansion of HSPCs, is associated with adiposity and future metabolic deterioration in healthy individuals.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Benedetta Maria Bonora
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Giorgio Marcuzzo
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Maria Cristina Marescotti
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Roberta Cappellari
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Giorgia Pantano
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Maria Colomba Sanzari
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Xavier Duran
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Joan Vendrell
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Mario Plebani
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| | - Angelo Avogaro
- Department of Medicine (G.P.F., B.M.B., M.C.M., R.C., G.P., M.C.S., M.P., A.A.), University of Padova, 35128 Padova, Italy; Venetian Institute of Molecular Medicine (G.P.F., X.D., A.A.), 35128 Padova, Italy; Department of Cardiologic, Thoracic and Vascular Sciences (G.M.), Service of Preventive Medicine, University of Padova, 35128 Padova, Italy; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) (X.D., J.V.), Instituto de Salud Carlos III, Madrid, Spain; Joan XXIII University Hospital (J.V.), Rovira i Virgili University IISPV, Tarragona, Spain
| |
Collapse
|
37
|
Luche E, Sengenès C, Arnaud E, Laharrague P, Casteilla L, Cousin B. Differential Hematopoietic Activity in White Adipose Tissue Depending on its Localization. J Cell Physiol 2015; 230:3076-83. [DOI: 10.1002/jcp.25045] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 05/12/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Elodie Luche
- CNRS UMR 5273; STROMALab; BP 84225; Toulouse France
- Université de Toulouse 3; UPS, BP 84225; Toulouse France
- INSERM U1031, BP 84225; Toulouse France
- EFS Pyrénées-Méditerranée, BP 84225; Toulouse France
| | - Coralie Sengenès
- CNRS UMR 5273; STROMALab; BP 84225; Toulouse France
- Université de Toulouse 3; UPS, BP 84225; Toulouse France
- INSERM U1031, BP 84225; Toulouse France
- EFS Pyrénées-Méditerranée, BP 84225; Toulouse France
| | - Emmanuelle Arnaud
- CNRS UMR 5273; STROMALab; BP 84225; Toulouse France
- Université de Toulouse 3; UPS, BP 84225; Toulouse France
- INSERM U1031, BP 84225; Toulouse France
- EFS Pyrénées-Méditerranée, BP 84225; Toulouse France
| | - Patrick Laharrague
- CNRS UMR 5273; STROMALab; BP 84225; Toulouse France
- Université de Toulouse 3; UPS, BP 84225; Toulouse France
- INSERM U1031, BP 84225; Toulouse France
- EFS Pyrénées-Méditerranée, BP 84225; Toulouse France
- Laboratoire d'Hématologie, TSA 50032; Toulouse France
| | - Louis Casteilla
- CNRS UMR 5273; STROMALab; BP 84225; Toulouse France
- Université de Toulouse 3; UPS, BP 84225; Toulouse France
- INSERM U1031, BP 84225; Toulouse France
- EFS Pyrénées-Méditerranée, BP 84225; Toulouse France
| | - Beatrice Cousin
- CNRS UMR 5273; STROMALab; BP 84225; Toulouse France
- Université de Toulouse 3; UPS, BP 84225; Toulouse France
- INSERM U1031, BP 84225; Toulouse France
- EFS Pyrénées-Méditerranée, BP 84225; Toulouse France
| |
Collapse
|
38
|
Bénézech C, Luu NT, Walker JA, Kruglov AA, Loo Y, Nakamura K, Zhang Y, Nayar S, Jones LH, Flores-Langarica A, McIntosh A, Marshall J, Barone F, Besra G, Miles K, Allen JE, Gray M, Kollias G, Cunningham AF, Withers DR, Toellner KM, Jones ND, Veldhoen M, Nedospasov SA, McKenzie ANJ, Caamaño JH. Inflammation-induced formation of fat-associated lymphoid clusters. Nat Immunol 2015; 16:819-828. [PMID: 26147686 PMCID: PMC4512620 DOI: 10.1038/ni.3215] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 06/01/2015] [Indexed: 12/14/2022]
Abstract
Fat-associated lymphoid clusters (FALCs) are a type of lymphoid tissue associated with visceral fat. Here we found that the distribution of FALCs was heterogeneous, with the pericardium containing large numbers of these clusters. FALCs contributed to the retention of B-1 cells in the peritoneal cavity through high expression of the chemokine CXCL13, and they supported B cell proliferation and germinal center differentiation during peritoneal immunological challenges. FALC formation was induced by inflammation, which triggered the recruitment of myeloid cells that expressed tumor-necrosis factor (TNF) necessary for signaling via the TNF receptors in stromal cells. Natural killer T cells (NKT cells) restricted by the antigen-presenting molecule CD1d were likewise required for the inducible formation of FALCs. Thus, FALCs supported and coordinated the activation of innate B cells and T cells during serosal immune responses.
Collapse
Affiliation(s)
- Cécile Bénézech
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Nguyet-Thin Luu
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | | | - Andrei A Kruglov
- German Rheumatism Research Center, Berlin, Germany
- Engelhardt Institute of Molecular Biology, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Yunhua Loo
- Lymphocyte Signalling and Development Programme, The Babraham Institute, Cambridge, UK
| | - Kyoko Nakamura
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Yang Zhang
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Saba Nayar
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Lucy H Jones
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh
| | - Adriana Flores-Langarica
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Alistair McIntosh
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Jennifer Marshall
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Francesca Barone
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Gurdyal Besra
- College of Life and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Katherine Miles
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Judith E Allen
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh
| | - Mohini Gray
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | | | - Adam F Cunningham
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - David R Withers
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Kai Michael Toellner
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Nick D Jones
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Marc Veldhoen
- Lymphocyte Signalling and Development Programme, The Babraham Institute, Cambridge, UK
| | - Sergei A Nedospasov
- German Rheumatism Research Center, Berlin, Germany
- Engelhardt Institute of Molecular Biology, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | | | - Jorge H Caamaño
- School of Immunity and Infection, IBR-MRC Centre for Immune Regulation, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
39
|
Abstract
Over the past decade, a series of discoveries relating to fibroblastic reticular cells (FRCs) — immunologically specialized myofibroblasts found in lymphoid tissue — has promoted these cells from benign bystanders to major players in the immune response. In this Review, we focus on recent advances regarding the immunobiology of lymph node-derived FRCs, presenting an updated view of crucial checkpoints during their development and their dynamic control of lymph node expansion and contraction during infection. We highlight the robust effects of FRCs on systemic B cell and T cell responses, and we present an emerging view of FRCs as drivers of pathology following acute and chronic viral infections. Lastly, we review emerging therapeutic advances that harness the immunoregulatory properties of FRCs.
Collapse
|
40
|
Differential Contributions of Graft-Derived and Host-Derived Cells in Tissue Regeneration/Remodeling after Fat Grafting. Plast Reconstr Surg 2015; 135:1607-1617. [DOI: 10.1097/prs.0000000000001292] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Abstract
Mesenchymal stromal cells (MSCs) are considered to be promising agents for the treatment of immunological disease. Although originally identified as precursor cells for mesenchymal lineages, in vitro studies have demonstrated that MSCs possess diverse immune regulatory capacities. Pre-clinical models have shown beneficial effects of MSCs in multiple immunological diseases and a number of phase 1/2 clinical trials carried out so far have reported signs of immune modulation after MSC infusion. These data indicate that MSCs play a central role in the immune response. This raises the academic question whether MSCs are immune cells or whether they are tissue precursor cells with immunoregulatory capacity. Correct understanding of the immunological properties and origin of MSCs will aid in the appropriate and safe use of the cells for clinical therapy. In this review the whole spectrum of immunological properties of MSCs is discussed with the aim of determining the position of MSCs in the immune system.
Collapse
Affiliation(s)
- Martin J Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, 3000, CA, Rotterdam, the Netherlands.
| |
Collapse
|
42
|
Bigot N, Guérillon C, Loisel S, Bertheuil N, Sensebé L, Tarte K, Pedeux R. ING1b negatively regulates HIF1α protein levels in adipose-derived stromal cells by a SUMOylation-dependent mechanism. Cell Death Dis 2015; 6:e1612. [PMID: 25611387 PMCID: PMC4669774 DOI: 10.1038/cddis.2014.577] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 11/18/2014] [Accepted: 12/03/2014] [Indexed: 12/16/2022]
Abstract
Hypoxic niches help maintain mesenchymal stromal cell properties, and their amplification under hypoxia sustains their immature state. However, how MSCs maintain their genomic integrity in this context remains elusive, since hypoxia may prevent proper DNA repair by downregulating expression of BRCA1 and RAD51. Here, we find that the ING1b tumor suppressor accumulates in adipose-derived stromal cells (ADSCs) upon genotoxic stress, owing to SUMOylation on K193 that is mediated by the E3 small ubiquitin-like modifier (SUMO) ligase protein inhibitor of activated STAT protein γ (PIAS4). We demonstrate that ING1b finely regulates the hypoxic response by triggering HIF1α proteasomal degradation. On the contrary, when mutated on its SUMOylation site, ING1b failed to efficiently decrease HIF1α levels. Consistently, we observed that the adipocyte differentiation, generally described to be downregulated by hypoxia, was highly dependent on ING1b expression, during the early days of this process. Accordingly, contrary to what was observed with HIF1α, the absence of ING1b impeded the adipogenic induction under hypoxic conditions. These data indicate that ING1b contributes to adipogenic induction in adipose-derived stromal cells, and thus hinders the phenotype maintenance of ADSCs.
Collapse
Affiliation(s)
- N Bigot
- 1] INSERM U917, Microenvironnement et Cancer, Rennes, France [2] Université de Rennes 1, Rennes, France [3] Etablissement Français du Sang Bretagne, Rennes, France
| | - C Guérillon
- 1] INSERM U917, Microenvironnement et Cancer, Rennes, France [2] Université de Rennes 1, Rennes, France [3] Etablissement Français du Sang Bretagne, Rennes, France
| | - S Loisel
- 1] INSERM U917, Microenvironnement et Cancer, Rennes, France [2] Université de Rennes 1, Rennes, France [3] Etablissement Français du Sang Bretagne, Rennes, France
| | - N Bertheuil
- 1] Université de Rennes 1, Rennes, France [2] Service ITeCH, CHU Pontchaillou, Rennes, France
| | - L Sensebé
- 1] Etablissement Français du Sang Pyrénées Méditerranée [2] Université Paul Sabatier, Toulouse, France [3] UMR5273-INSERM U1031, Toulouse, France
| | - K Tarte
- 1] INSERM U917, Microenvironnement et Cancer, Rennes, France [2] Université de Rennes 1, Rennes, France [3] Etablissement Français du Sang Bretagne, Rennes, France [4] Service ITeCH, CHU Pontchaillou, Rennes, France
| | - R Pedeux
- 1] INSERM U917, Microenvironnement et Cancer, Rennes, France [2] Université de Rennes 1, Rennes, France [3] Etablissement Français du Sang Bretagne, Rennes, France
| |
Collapse
|
43
|
Hoogduijn MJ, Verstegen MM, Engela AU, Korevaar SS, Roemeling-van Rhijn M, Merino A, Franquesa M, de Jonge J, Ijzermans JN, Weimar W, Betjes MG, Baan CC, van der Laan LJ. No Evidence for Circulating Mesenchymal Stem Cells in Patients with Organ Injury. Stem Cells Dev 2014; 23:2328-35. [DOI: 10.1089/scd.2014.0269] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Martin J. Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | - Anja U. Engela
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Sander S. Korevaar
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | - Ana Merino
- Department of Experimental Nephrology, Bellvitge University Hospital, Barcelona, Spain
| | - Marcella Franquesa
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Jeroen de Jonge
- Department of Surgery, Erasmus MC, Rotterdam, The Netherlands
| | | | - Willem Weimar
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Michiel G.H. Betjes
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Carla C. Baan
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | |
Collapse
|
44
|
Eggenhofer E, Luk F, Dahlke MH, Hoogduijn MJ. The life and fate of mesenchymal stem cells. Front Immunol 2014; 5:148. [PMID: 24904568 PMCID: PMC4032901 DOI: 10.3389/fimmu.2014.00148] [Citation(s) in RCA: 342] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 03/21/2014] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSC) are present throughout the body and are thought to play a role in tissue regeneration and control of inflammation. MSC can be easily expanded in vitro and their potential as a therapeutic option for degenerative and inflammatory disease is therefore intensively investigated. Whilst it was initially thought that MSC would replace dysfunctional cells and migrate to sites of injury to interact with inflammatory cells, experimental evidence indicates that the majority of administered MSC get trapped in capillary networks and have a short life span. In this review, we discuss current knowledge on the migratory properties of endogenous and exogenous MSC and confer on how culture-induced modifications of MSC may affect these properties. Finally, we will discuss how, despite their limited survival, administered MSC can bring about their therapeutic effects.
Collapse
Affiliation(s)
- Elke Eggenhofer
- Department of Surgery, University Medical Center Regensburg , Regensburg , Germany
| | - Franka Luk
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center , Rotterdam , Netherlands
| | - Marc H Dahlke
- Department of Surgery, University Medical Center Regensburg , Regensburg , Germany
| | - Martin J Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center , Rotterdam , Netherlands
| |
Collapse
|
45
|
Heyboer M, Milovanova TN, Wojcik S, Grant W, Chin M, Hardy KR, Lambert DS, Logue C, Thom SR. CD34+/CD45-dim stem cell mobilization by hyperbaric oxygen - changes with oxygen dosage. Stem Cell Res 2014; 12:638-45. [PMID: 24642336 PMCID: PMC4037447 DOI: 10.1016/j.scr.2014.02.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/14/2014] [Accepted: 02/22/2014] [Indexed: 11/16/2022] Open
Abstract
Because hyperbaric oxygen treatment mobilizes bone marrow derived-stem/progenitor cells by a free radical mediated mechanism, we hypothesized that there may be differences in mobilization efficiency based on exposure to different oxygen partial pressures. Blood from twenty consecutive patients was obtained before and after the 1st, 10th and 20th treatment at two clinical centers using protocols involving exposures to oxygen at either 2.0 or 2.5 atmospheres absolute (ATA). Post-treatment values of CD34+, CD45-dim leukocytes were always 2-fold greater than the pre-treatment values for both protocols. Values for those treated at 2.5 ATA were significantly greater than those treated at 2.0 ATA by factors of 1.9 to 3-fold after the 10th and before and after the 20th treatments. Intracellular content of hypoxia inducible factors -1, -2, and -3, thioredoxin-1 and poly-ADP-ribose polymerase assessed in permeabilized CD34+ cells with fluorophore-conjugated antibodies were twice as high in all post- versus pre-treatment samples with no significant differences between 2.0 and 2.5 ATA protocols. We conclude that putative progenitor cell mobilization is higher with 2.5 versus 2.0 ATA treatments, and all newly mobilized cells exhibit higher concentrations of an array of regulatory proteins.
Collapse
Affiliation(s)
- Marvin Heyboer
- Department of Emergency Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Tatyana N Milovanova
- Institute for Environmental Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Susan Wojcik
- Department of Emergency Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - William Grant
- Department of Emergency Medicine, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Mary Chin
- Institute for Environmental Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kevin R Hardy
- Institute for Environmental Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David S Lambert
- Institute for Environmental Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher Logue
- Institute for Environmental Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen R Thom
- Institute for Environmental Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
46
|
Gil-Ortega M, Fernández-Alfonso MS, Somoza B, Casteilla L, Sengenès C. Ex vivo microperfusion system of the adipose organ: a new approach to studying the mobilization of adipose cell populations. Int J Obes (Lond) 2013; 38:1255-62. [PMID: 24357852 DOI: 10.1038/ijo.2013.243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 11/28/2013] [Accepted: 12/16/2013] [Indexed: 12/26/2022]
Abstract
BACKGROUND/OBJECTIVES Adipose tissue (AT) is a dynamic organ that expands and contracts rapidly. It is composed of adipocytes and of cell populations among which immune cells and mesenchymal progenitors known as adipose stromal cells (ASCs). The AT cell turnover has been extensively studied. Surprisingly it has only been viewed as the result of both cell proliferation/death and cell infiltration. Nevertheless, both immune cells and ASCs exhibit migration abilities; therefore their egress from AT in response to physiological/pathophysiological stimuli has to be considered. To do so, the aim of the present work was to develop a model allowing the study of cell release from the adipose organ. SUBJECTS/METHODS Mesenteric (Mes) ATs were isolated from 9-week-old C57BL/6 male mice and were catheterized via the superior mesenteric artery and were perfused with a saline solution. After an equilibration period, the mesenteric fat pad was perfused with CXCL12 (stromal-derived factor-1, SDF-1) or sphingosine 1-phosphate (S1P) to trigger cell mobilization and perfusates were collected every 30 min for subsequent flow cytometry analyses. RESULTS We report here that CXCL12 induces the specific release of ASCs from MesAT thus demonstrating that ASCs are specifically mobilized from fat depots by a CXCL12-dependent pathway. Moreover, we showed that leukocyte mobilization can be triggered via a S1P-dependent pathway. CONCLUSIONS We have developed a microperfusion model of an intact fat depot allowing the study of AT cell release in response to various molecules. The perfusion system described here demonstrates that ASCs and leukocytes can be pharmacologically mobilized from AT. Therefore, AT microperfusion might constitute an appropriate and reliable approach for evaluating the mobilization of different cell populations from AT in various physiological and pathophysiological contexts. Such a model might help in identifying factors and drugs controlling AT cell release, impacting the medical fields of regenerative medicine and of obesity or its associated comorbidities.
Collapse
Affiliation(s)
- M Gil-Ortega
- 1] Inserm U1031 STROMAlab BP 84 225-F-31 432, Toulouse, France [2] CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, BP 84 225-F-31 432, Toulouse, France [3] EFS (Etablissement Français du Sang), STROMAlab BP 84 225-F-31 432, Toulouse, France [4] Université Toulouse III, UPS UMR5273 STROMAlab BP 84 225-F-31 432, Toulouse, France
| | - M S Fernández-Alfonso
- Instituto Pluridisciplinar, Facultad de Farmacia, Universidad Complutense de Madrid, Juan XXIII 1, 28040 Madrid, Spain
| | - B Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, 28668 Madrid, Spain
| | - L Casteilla
- 1] Inserm U1031 STROMAlab BP 84 225-F-31 432, Toulouse, France [2] CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, BP 84 225-F-31 432, Toulouse, France [3] EFS (Etablissement Français du Sang), STROMAlab BP 84 225-F-31 432, Toulouse, France [4] Université Toulouse III, UPS UMR5273 STROMAlab BP 84 225-F-31 432, Toulouse, France
| | - C Sengenès
- 1] Inserm U1031 STROMAlab BP 84 225-F-31 432, Toulouse, France [2] CNRS, Université Toulouse III, UPS UMR5273 STROMAlab, BP 84 225-F-31 432, Toulouse, France [3] EFS (Etablissement Français du Sang), STROMAlab BP 84 225-F-31 432, Toulouse, France [4] Université Toulouse III, UPS UMR5273 STROMAlab BP 84 225-F-31 432, Toulouse, France
| |
Collapse
|