1
|
Ziadlou R, Pandian GN, Hafner J, Akdis CA, Stingl G, Maverakis E, Brüggen M. Subcutaneous adipose tissue: Implications in dermatological diseases and beyond. Allergy 2024; 79:3310-3325. [PMID: 39206504 PMCID: PMC11657049 DOI: 10.1111/all.16295] [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: 02/08/2024] [Revised: 07/19/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Subcutaneous adipose tissue (SAT) is the deepest component of the three-layered cutaneous integument. While mesenteric adipose tissue-based immune processes have gained recognition in the context of the metabolic syndrome, SAT has been traditionally considered primarily for energy storage, with less attention to its immune functions. SAT harbors a reservoir of immune and stromal cells that significantly impact metabolic and immunologic processes not only in the skin, but even on a systemic level. These processes include wound healing, cutaneous and systemic infections, immunometabolic, and autoimmune diseases, inflammatory skin diseases, as well as neoplastic conditions. A better understanding of SAT immune functions in different processes, could open avenues for novel therapeutic interventions. Targeting SAT may not only address SAT-specific diseases but also offer potential treatments for cutaneous or even systemic conditions. This review aims to provide a comprehensive overview on SAT's structure and functions, highlight recent advancements in understanding its role in both homeostatic and pathological conditions within and beyond the skin, and discuss the main questions for future research in the field.
Collapse
Affiliation(s)
- Reihane Ziadlou
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
- Christine Kühne Center for Allergy Research and Education CK‐CAREDavosSwitzerland
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichZurichSwitzerland
| | - Ganesh N. Pandian
- Institute for Integrated Cell‐Material Science (WPI‐iCeMS)Kyoto UniversityKyotoJapan
| | - Jürg Hafner
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
| | - Cezmi A. Akdis
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
- Christine Kühne Center for Allergy Research and Education CK‐CAREDavosSwitzerland
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichZurichSwitzerland
| | - Georg Stingl
- Department of DermatologyMedical University of ViennaViennaAustria
| | | | - Marie‐Charlotte Brüggen
- Faculty of MedicineUniversity of ZurichZurichSwitzerland
- Department of DermatologyUniversity Hospital ZurichZurichSwitzerland
- Christine Kühne Center for Allergy Research and Education CK‐CAREDavosSwitzerland
| |
Collapse
|
2
|
Tong X, Zhu Q, Duo T, Liang Z, Zhang C, Cai S, Wang X, Liu Y, Li Y, Liu X, He Z, Hu B, Zeng J, Chen Y, Mo D. The Impact of FBN1-α5β1 Axis in Fibro/Adipogenic Progenitor Cells (FAP CD9-) on Intramuscular Fat Content in Pigs. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 38598771 DOI: 10.1021/acs.jafc.4c00059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Intramuscular fat (IMF) plays a crucial role in enhancing meat quality, enriching meat flavor, and overall improving palatability. In this study, Single-cell RNA sequencing was employed to analyze the longissimus dorsi (LD) obtained from Guangdong small-ear spotted pigs (GDSS, with high IMF) and Yorkshire pigs (YK, with low IMF). GDSS had significantly more Fibro/Adipogenic Progenitor (FAPs), in which the CD9 negative FAPs (FAPCD9-) having adipogenic potential, as demonstrated by in vitro assays using cells originated from mouse muscle. On the other hand, Yorkshire had more fibro-inflammatory progenitors (FIPs, marked with FAPCD9+), presenting higher expression of the FBN1-Integrin α5β1. FBN1-Integrin α5β1 could inhibit insulin signaling in FAPCD9-, suppressing adipogenic differentiation. Our results demonstrated that fat-type pigs possess a greater number of FAPCD9-, which are the exclusive cells in muscle capable of differentiating into adipocytes. Moreover, lean-type pigs exhibit higher expression of FBN1-Integrin α5β1 axis, which inhibits adipocyte differentiation. These results appropriately explain the observed higher IMF content in fat-type pigs.
Collapse
Affiliation(s)
- Xian Tong
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Qi Zhu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Tianqi Duo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Ziyun Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Chong Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Shufang Cai
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Xiaoyu Wang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Yihao Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Yongpeng Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Zuyong He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Bin Hu
- Guangdong Key Laboratory of Animal Breeding and Nutrition, State Key, Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong 510640, China
| | - Jianhua Zeng
- Guangdong YIHAO Food Co.,Ltd., Guangzhou 510620, China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, China
| |
Collapse
|
3
|
Picoli CDC, Birbrair A, Li Z. Pericytes as the Orchestrators of Vasculature and Adipogenesis. Genes (Basel) 2024; 15:126. [PMID: 38275607 PMCID: PMC10815550 DOI: 10.3390/genes15010126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Pericytes (PCs) are located surrounding the walls of small blood vessels, particularly capillaries and microvessels. In addition to their functions in maintaining vascular integrity, participating in angiogenesis, and regulating blood flow, PCs also serve as a reservoir for multi-potent stem/progenitor cells in white, brown, beige, and bone marrow adipose tissues. Due to the complex nature of this cell population, the identification and characterization of PCs has been challenging. A comprehensive understanding of the heterogeneity of PCs may enhance their potential as therapeutic targets for metabolic syndromes or bone-related diseases. This mini-review summarizes multiple PC markers commonly employed in lineage-tracing studies, with an emphasis on their contribution to adipogenesis and functions in different adipose depots under diverse metabolic conditions.
Collapse
Affiliation(s)
| | - Alexander Birbrair
- Department of Dermatology, University of Wisconsin-Madison, Medical Sciences Center, Madison, WI 53706, USA;
| | - Ziru Li
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME 04074, USA;
| |
Collapse
|
4
|
Ernst AM, Steiner M, Kainz V, Tempfer H, Spitzer G, Plank T, Bauer HC, Bresgen N, Habenbacher A, Bauer H, Lipp AT. Lipedema: The Use of Cultured Adipocytes for Identification of Diagnostic Markers. Plast Reconstr Surg 2023; 152:1036-1046. [PMID: 36912938 DOI: 10.1097/prs.0000000000010392] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
BACKGROUND Lipedema, diagnosed most often in women, is a progressive disease characterized by the disproportionate and symmetrical distribution of adipose tissue, primarily in the extremities. Although numerous results from in vitro and in vivo studies have been published, many questions regarding the pathology and genetic background of lipedema remain unanswered. METHODS In this study, adipose tissue-derived stromal/stem cells were isolated from lipoaspirates derived from nonobese and obese donors with or without lipedema. Growth and morphology, metabolic activity, differentiation potential, and gene expression were evaluated using quantification of lipid accumulation, metabolic activity assay, live-cell imaging, reverse transcription polymerase chain reaction, quantitative polymerase chain reaction, and immunocytochemical staining. RESULTS The adipogenic potential of lipedema and nonlipedema adipose tissue-derived stromal/stem cells did not rise in parallel with the donors' body mass index and did not differ significantly between groups. However, in vitro differentiated adipocytes from nonobese lipedema donors showed significant upregulation of adipogenic gene expression compared with nonobese controls. All other genes tested were expressed equally in lipedema and nonlipedema adipocytes. The adiponectin/leptin ratio was significantly reduced in adipocytes from obese lipedema donors compared with their nonobese lipedema counterparts. Increased stress fiber-integrated smooth muscle actin was visible in lipedema adipocytes compared with nonlipedema controls and appeared enhanced in adipocytes from obese lipedema donors. CONCLUSIONS Not only lipedema per se but also body mass index of donors affect adipogenic gene expression substantially in vitro. The significantly reduced adiponectin/leptin ratio and the increased occurrence of myofibroblast-like cells in obese lipedema adipocyte cultures underscores the importance of attention to the co-occurrence of lipedema and obesity. These are important findings toward accurate diagnosis of lipedema. CLINICAL RELEVANCE STATEMENT Our study highlights not only the difficulty in lipedema diagnostics but also the tremendous need for further studies on lipedema tissue. Although lipedema might seem to be an underestimated field in plastic and reconstructive surgery, the power it holds to provide better treatment to future patients can not be promoted enough.
Collapse
Affiliation(s)
- Anna M Ernst
- From the Department of Biosciences, Paris Lodron University of Salzburg
| | - Marianne Steiner
- From the Department of Biosciences, Paris Lodron University of Salzburg
| | - Verena Kainz
- From the Department of Biosciences, Paris Lodron University of Salzburg
| | - Herbert Tempfer
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration Centre Salzburg, Paracelsus Medical University Salzburg
- Austrian Cluster for Tissue Regeneration
| | - Gabriel Spitzer
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration Centre Salzburg, Paracelsus Medical University Salzburg
- Austrian Cluster for Tissue Regeneration
| | - Tanja Plank
- From the Department of Biosciences, Paris Lodron University of Salzburg
| | - Hans-Christian Bauer
- From the Department of Biosciences, Paris Lodron University of Salzburg
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration Centre Salzburg, Paracelsus Medical University Salzburg
- Austrian Cluster for Tissue Regeneration
| | - Nikolaus Bresgen
- From the Department of Biosciences, Paris Lodron University of Salzburg
| | - Andreas Habenbacher
- Department of Plastic Surgery and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich
| | - Hannelore Bauer
- From the Department of Biosciences, Paris Lodron University of Salzburg
| | - Anna-Theresa Lipp
- Department of Plastic Surgery and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich
| |
Collapse
|
5
|
Song W, Liu P, Li H, Ding S. Large-Scale Expansion of Porcine Adipose-Derived Stem Cells Based on Microcarriers System for Cultured Meat Production. Foods 2022; 11:foods11213364. [PMID: 36359977 PMCID: PMC9656844 DOI: 10.3390/foods11213364] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 12/02/2022] Open
Abstract
Cultured meat is an innovative meat-production technology that does not rely on animal husbandry. As a new food component, cultured fat is of great significance to cultured meat. In this study, we isolated adipose-derived stem cells (ADSCs) and identified the purity by immunofluorescence staining of ADSC-specific surface marker proteins CD44 and CD29 and showed that most of the cells were positive for CD29 and CD44. In addition, we detected the expression of FABP4 and Plin1 to confirm that ADSCs differentiated into mature adipocytes at 10 days post-induction. Subsequently, the culture conditions of ADSCs on microcarriers (MCs) were optimized and showed that cell density of living cells reached their highest after 5 days when continuously stirring at 50 rpm. Finally, the expression of FABP4 and PPARγ was detected to confirm the adipogenic differentiation ability of ADSCs on 2D and 3D culture systems and showed that ADSCs maintained their adipogenic differentiation ability after expansion on MCs. In conclusion, this research demonstrated that reliance on MCs to expand ADSCs was a promising approach for production of cultured fat.
Collapse
Affiliation(s)
- Wenjuan Song
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Peipei Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Huixia Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- Correspondence: (H.L.); (S.D.)
| | - Shijie Ding
- College of Food Science and Technology, Nanjing Agricultural University, National Center of Meat Quality and Safety Nanjing, Nanjing 210095, China
- Correspondence: (H.L.); (S.D.)
| |
Collapse
|
6
|
Kislev N, Izgilov R, Adler R, Benayahu D. Exploring the Cell Stemness and the Complexity of the Adipose Tissue Niche. Biomolecules 2021; 11:biom11121906. [PMID: 34944549 PMCID: PMC8699211 DOI: 10.3390/biom11121906] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/05/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is a complex organ composed of different cellular populations, including mesenchymal stem and progenitor cells, adipocytes, and immune cells such as macrophages and lymphocytes. These cellular populations alter dynamically during aging or as a response to pathophysiology such as obesity. Changes in the various inflammatory cells are associated with metabolic complications and the development of insulin resistance, indicating that immune cells crosstalk with the adipocytes. Therefore, a study of the cell populations in the adipose tissue and the extracellular matrix maintaining the tissue niche is important for the knowledge on the regulatory state of the organ. We used a combination of methods to study various parameters to identify the composition of the resident cells in the adipose tissue and evaluate their profile. We analyzed the tissue structure and cells based on histology, immune fluorescence staining, and flow cytometry of cells present in the tissue in vivo and these markers’ expression in vitro. Any shift in cells’ composition influences self-renewal of the mesenchymal progenitors, and other cells affect the functionality of adipogenesis.
Collapse
|
7
|
Abstract
Expansion of visceral white adipose tissue (vWAT) occurs in response to nutrient excess, and is a risk factor for metabolic disease. SPRY1, a feedback inhibitor of receptor tyrosine kinase (RTK) signaling, is expressed in PDGFRa+ adipocyte progenitor cells (APC) in vivo. Global deficiency of Spry1 in mice results in disproportionate postnatal growth of gonadal WAT (gWAT), while iWAT and BAT were similar in size between Spry1KO and WT mice. Spry1 deficiency increased the number of PDGFRa+ stromal vascular fraction (SVF) cells in gWAT and showed increased proliferation and fibrosis. Spry1KO gWAT had increased collagen deposition and elevated expression of markers of inflammation. In vitro, SPRY1 was transiently down regulated during early adipocyte differentiation of SVF cells, with levels increasing at later stages of differentiation. SPRY1 deficiency enhances PDGF-AA and PDGF-BB induced proliferation of SVF cells. Increased proliferation of SVF from Spry1KO gWAT accompanies an increase in AKT activation. PDGF-AA stimulated a transient down regulation of SPRY1 in wild type SVF, whereas PDGF-BB stimulated a sustained down regulation of SPRY1 in wild type SVF. Collectively, our data suggest that SPRY1 is critical for regulating postnatal growth of gWAT by restraining APC proliferation and differentiation in part by regulation of PDGFRa/b-AKT signaling.
Collapse
Affiliation(s)
- Xuehui Yang
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Shivangi Pande
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| | - Robert A. Koza
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| | - Robert Friesel
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| |
Collapse
|
8
|
Adult Human Multipotent Neural Cells Could Be Distinguished from Other Cell Types by Proangiogenic Paracrine Effects via MCP-1 and GRO. Stem Cells Int 2021; 2021:6737288. [PMID: 34434240 PMCID: PMC8380502 DOI: 10.1155/2021/6737288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/28/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Adult human multipotent neural cells (ahMNCs) are unique cells derived from adult human temporal lobes. They show multipotent differentiation potentials into neurons and astrocytes. In addition, they possess proangiogenic capacities. The objective of this study was to characterize ahMNCs in terms of expression of cell type-specific markers, in vitro differentiation potentials, and paracrine factors compared with several other cell types including fetal neural stem cells (fNSCs) to provide detailed molecular and functional features of ahMNCs. Interestingly, the expression of cell type-specific markers of ahMNCs could not be differentiated from those of pericytes, mesenchymal stem cells (MSCs), or fNSCs. In contrast, differentiation potentials of ahMNCs and fNSCs into neural cells were higher than those of other cell types. Compared with MSCs, ahMNCs showed lower differentiation capacities into osteogenic and adipogenic cells. Moreover, ahMNCs uniquely expressed higher levels of MCP-1 and GRO family paracrine factors than fNSCs and MSCs. These high levels of MCP-1 and GRO family mediated in vivo proangiogenic effects of ahMNCs. These results indicate that ahMNCs have their own distinct characteristics that could distinguish ahMNCs from other cell types. Characteristics of ahMNCs could be utilized further in the preclinical and clinical development of ahMNCs for regenerative medicine. They could also be used as experimental references for other cell types including fNSCs.
Collapse
|
9
|
Garita B, Maligro J, Sadoughi S, Wu PH, Liebenberg E, Horvai A, Link TM, Kazakia GJ. Microstructural abnormalities are evident by histology but not HR-pQCT at the periosteal cortex of the human tibia under CVD and T2D conditions. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2021. [DOI: 10.1016/j.medntd.2021.100062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
10
|
Porro S, Genchi VA, Cignarelli A, Natalicchio A, Laviola L, Giorgino F, Perrini S. Dysmetabolic adipose tissue in obesity: morphological and functional characteristics of adipose stem cells and mature adipocytes in healthy and unhealthy obese subjects. J Endocrinol Invest 2021; 44:921-941. [PMID: 33145726 DOI: 10.1007/s40618-020-01446-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
The way by which subcutaneous adipose tissue (SAT) expands and undergoes remodeling by storing excess lipids through expansion of adipocytes (hypertrophy) or recruitment of new precursor cells (hyperplasia) impacts the risk of developing cardiometabolic and respiratory diseases. In unhealthy obese subjects, insulin resistance, type 2 diabetes, hypertension, and obstructive sleep apnoea are typically associated with pathologic SAT remodeling characterized by adipocyte hypertrophy, as well as chronic inflammation, hypoxia, increased visceral adipose tissue (VAT), and fatty liver. In contrast, metabolically healthy obese individuals are generally associated with SAT development characterized by the presence of smaller and numerous mature adipocytes, and a lower degree of VAT inflammation and ectopic fat accumulation. The remodeling of SAT and VAT is under genetic regulation and influenced by inherent depot-specific differences of adipose tissue-derived stem cells (ASCs). ASCs have multiple functions such as cell renewal, adipogenic capacity, and angiogenic properties, and secrete a variety of bioactive molecules involved in vascular and extracellular matrix remodeling. Understanding the mechanisms regulating the proliferative and adipogenic capacity of ASCs from SAT and VAT in response to excess calorie intake has become a focus of interest over recent decades. Here, we summarize current knowledge about the biological mechanisms able to foster or impair the recruitment and adipogenic differentiation of ASCs during SAT and VAT development, which regulate body fat distribution and favorable or unfavorable metabolic responses.
Collapse
Affiliation(s)
- S Porro
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - V A Genchi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Cignarelli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Natalicchio
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - L Laviola
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - F Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy.
| | - S Perrini
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| |
Collapse
|
11
|
Girolamo F, de Trizio I, Errede M, Longo G, d'Amati A, Virgintino D. Neural crest cell-derived pericytes act as pro-angiogenic cells in human neocortex development and gliomas. Fluids Barriers CNS 2021; 18:14. [PMID: 33743764 PMCID: PMC7980348 DOI: 10.1186/s12987-021-00242-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Central nervous system diseases involving the parenchymal microvessels are frequently associated with a ‘microvasculopathy’, which includes different levels of neurovascular unit (NVU) dysfunction, including blood–brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches. ![]()
Collapse
Affiliation(s)
- Francesco Girolamo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.
| | - Ignazio de Trizio
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Intensive Care Unit, Department of Intensive Care, Regional Hospital of Lugano, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Mariella Errede
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| | - Giovanna Longo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Molecular Biology Unit, University of Bari School of Medicine, Bari, Italy
| | - Antonio d'Amati
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy.,Department of Emergency and Organ Transplantation, Pathology Section, University of Bari School of Medicine, Bari, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Human Anatomy and Histology Unit, University of Bari School of Medicine, Bari, Italy
| |
Collapse
|
12
|
Girolamo F, de Trizio I, Errede M, Longo G, d’Amati A, Virgintino D. Neural crest cell-derived pericytes act as pro-angiogenic cells in human neocortex development and gliomas. Fluids Barriers CNS 2021. [DOI: 10.1186/s12987-021-00242-7 union select null--] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractCentral nervous system diseases involving the parenchymal microvessels are frequently associated with a ‘microvasculopathy’, which includes different levels of neurovascular unit (NVU) dysfunction, including blood–brain barrier alterations. To contribute to the understanding of NVU responses to pathological noxae, we have focused on one of its cellular components, the microvascular pericytes, highlighting unique features of brain pericytes with the aid of the analyses carried out during vascularization of human developing neocortex and in human gliomas. Thanks to their position, centred within the endothelial/glial partition of the vessel basal lamina and therefore inserted between endothelial cells and the perivascular and vessel-associated components (astrocytes, oligodendrocyte precursor cells (OPCs)/NG2-glia, microglia, macrophages, nerve terminals), pericytes fulfil a central role within the microvessel NVU. Indeed, at this critical site, pericytes have a number of direct and extracellular matrix molecule- and soluble factor-mediated functions, displaying marked phenotypical and functional heterogeneity and carrying out multitasking services. This pericytes heterogeneity is primarily linked to their position in specific tissue and organ microenvironments and, most importantly, to their ontogeny. During ontogenesis, pericyte subtypes belong to two main embryonic germ layers, mesoderm and (neuro)ectoderm, and are therefore expected to be found in organs ontogenetically different, nonetheless, pericytes of different origin may converge and colonize neighbouring areas of the same organ/apparatus. Here, we provide a brief overview of the unusual roles played by forebrain pericytes in the processes of angiogenesis and barriergenesis by virtue of their origin from midbrain neural crest stem cells. A better knowledge of the ontogenetic subpopulations may support the understanding of specific interactions and mechanisms involved in pericyte function/dysfunction, including normal and pathological angiogenesis, thereby offering an alternative perspective on cell subtype-specific therapeutic approaches.
Collapse
|
13
|
Invited Discussion on: Effects of Harvest Sites on Cryopreserved Adipose-Derived Stem Cells and ASC-Enriched Fat Grafts. Aesthetic Plast Surg 2020; 44:2297-2299. [PMID: 32940734 DOI: 10.1007/s00266-020-01951-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 01/22/2023]
|
14
|
Zhang Z, Paudel S, Feltham T, Lobao MH, Schon L. Foot fat pad: Characterization by mesenchymal stromal cells in rats. Anat Rec (Hoboken) 2020; 304:1582-1591. [PMID: 33099882 DOI: 10.1002/ar.24549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/02/2020] [Accepted: 09/14/2020] [Indexed: 11/10/2022]
Abstract
Foot fat pad (FFP) is a highly functionalized fat depot of great significance for weight bearing in the foot. Mesenchymal stromal cells (MSCs) in subcutaneous adipose tissues are widely studied for regenerative potentials. MSCs in FFP, which may contribute to the physiological and pathological conditions of the foot, have not been characterized. In this study, MSCs were isolated from FFP (designated as MSCs-ffp) and subcutaneous adipose tissue (designated as MSCs-sub) from rats. The cell surface markers, proliferation, and efficiency of colony formation were compared between MSCs-ffp and MSCs-sub. In addition, MSCs-ffp were induced for osteogenic, chondrogenic, and adipogenic differentiation. The tri-lineage differentiation potentials were compared between MSCs-ffp and MSCs-sub by the expression of Runx2, Sox9, and proliferator-activated receptor gamma (PPAR-γ), respectively, using quantitative polymerized chain reaction. The expression of elastin and associated genes by MSCs-ffp were also evaluated. MSCs-ffp, like MSCs-sub, expressed CD44, CD73, and CD90. MSCs-ffp and MSCs-sub proliferated at similar rates but MSCs-ffp formed more colonies than MSCs-sub. MSCs-ffp were capable of differentiating into osteogenic, chondrogenic, and adipogenic lineages. Under the conditions of osteogenic and adipogenic differentiation, MSCs-sub expressed more Runx2 and PPAR-γ, respectively, than MSCs-ffp. The undifferentiated MSCs-ffp upregulated the expression of fibulin-5. In conclusion, MSCs-ffp shared common biology with MSCs-sub but were more efficient in colony formation, less adipogenic and osteogenic, and participated in elastogenesis. The unique features of MSCs-ffp may relate to their roles in the physiological functions of FFP.
Collapse
Affiliation(s)
- Zijun Zhang
- Center for Orthopaedic Innovation, Mercy Medical Center, Baltimore, Maryland, USA
| | - Sharada Paudel
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Tyler Feltham
- Philadelphia College of Osteopathic Medicine-GA, Suwanee, Georgia, USA
| | - Mario H Lobao
- Department of Orthopaedic Surgery, Columbia University Medical Center, New York, New York, USA
| | - Lew Schon
- Center for Orthopaedic Innovation, Mercy Medical Center, Baltimore, Maryland, USA.,Institute for Foot and Ankle Reconstruction, Mercy Medical Center, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Hussain A, Deleuze V, El Kebriti L, Turali H, Pirot N, Glasson Y, Mathieu D, Pinet V. In Lyl1 -/- mice, adipose stem cell vascular niche impairment leads to premature development of fat tissues. STEM CELLS (DAYTON, OHIO) 2020; 39:78-91. [PMID: 33022858 PMCID: PMC7821250 DOI: 10.1002/stem.3286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022]
Abstract
Lyl1 encodes a hematopoietic‐ and endothelial‐specific bHLH transcription factor. Lyl1‐deficient mice are viable, but they display mild hematopoietic and vascular defects. Specifically, LYL1 is required for the maturation and stabilization of blood vessel endothelial adherens junctions. Here, we report that young adult Lyl1−/− mice exhibit transient overweight associated with general expansion of adipose tissue, without signs of metabolic disorder and unrelated to food intake. The increased fat tissue development in Lyl1−/− mice resulted from earlier differentiation of adipose stem cells (ASCs) into adipocytes through noncell autonomous mechanisms. Specifically, we found that in Lyl1−/− mice, the adipose tissue vascular structures are immature, as indicated by their high permeability, reduced coverage by pericytes, lower recruitment of VE‐cadherin and ZO1 at cell junctions, and more prone to angiogenesis. Together, our data show that in Lyl1−/− mice, the impaired vascular compartment of the adipose niche promotes ASC differentiation, leading to early adipocyte expansion and premature ASC depletion. Our study highlights the major structural role of the adipose tissue vascular niche in coordinating stem cell self‐renewal and differentiation into adipocytes.
Collapse
Affiliation(s)
- Abid Hussain
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Virginie Deleuze
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Leila El Kebriti
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Hulya Turali
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Nelly Pirot
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Yaël Glasson
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Danièle Mathieu
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Valérie Pinet
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| |
Collapse
|
16
|
Kim HW, Shi H, Winkler MA, Lee R, Weintraub NL. Perivascular Adipose Tissue and Vascular Perturbation/Atherosclerosis. Arterioscler Thromb Vasc Biol 2020; 40:2569-2576. [PMID: 32878476 DOI: 10.1161/atvbaha.120.312470] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Atherosclerosis is orchestrated by complex interactions between vascular and inflammatory cells. Traditionally, it has been considered to be an intimal inflammatory disease, characterized by endothelial dysfunction, inflammatory cell recruitment, lipid oxidation, and foam cell formation. This inside-out signaling paradigm has been accepted as dogma for many years, despite the fact that inflammatory cells are far more prevalent in the adventitia compared with the intima. For decades, the origin of adventitial inflammation in atherosclerosis was unknown. The fact that these inflammatory cells were observed to cluster at the margin of perivascular adipose tissues-a unique and highly inflammatory adipose depot that surrounds most atherosclerosis-prone blood vessels-has stimulated interest in perivascular adipose tissue-mediated outside-in signaling in vascular pathophysiology, including atherosclerosis. The phenotype of perivascular adipocytes underlies the functional characteristics of this depot, including its role in adventitial inflammatory cell recruitment, trafficking to the intima via the vasa vasorum, and atherosclerosis perturbation. This review is focused on emerging concepts pertaining to outside-in signaling in atherosclerosis driven by dysfunctional perivascular adipose tissues during diet-induced obesity and recent strategies for atherosclerosis prediction and prognostication based upon this hypothesis.
Collapse
Affiliation(s)
- Ha Won Kim
- Department of Medicine (H.W.K., H.S., N.L.W.), Medical College of Georgia at Augusta University.,Vascular Biology Center (H.W.K., H.S., N.L.W.), Medical College of Georgia at Augusta University
| | - Hong Shi
- Department of Medicine (H.W.K., H.S., N.L.W.), Medical College of Georgia at Augusta University.,Vascular Biology Center (H.W.K., H.S., N.L.W.), Medical College of Georgia at Augusta University
| | - Michael A Winkler
- Department of Radiology (M.A.W.), Medical College of Georgia at Augusta University
| | - Richard Lee
- Department of Surgery (R.L.), Medical College of Georgia at Augusta University
| | - Neal L Weintraub
- Department of Medicine (H.W.K., H.S., N.L.W.), Medical College of Georgia at Augusta University.,Vascular Biology Center (H.W.K., H.S., N.L.W.), Medical College of Georgia at Augusta University
| |
Collapse
|
17
|
Brzoska E, Kalkowski L, Kowalski K, Michalski P, Kowalczyk P, Mierzejewski B, Walczak P, Ciemerych MA, Janowski M. Muscular Contribution to Adolescent Idiopathic Scoliosis from the Perspective of Stem Cell-Based Regenerative Medicine. Stem Cells Dev 2020; 28:1059-1077. [PMID: 31170887 DOI: 10.1089/scd.2019.0073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is a relatively frequent disease within a range 0.5%-5.0% of population, with higher frequency in females. While a resultant spinal deformity is usually medically benign condition, it produces far going psychosocial consequences, which warrant attention. The etiology of AIS is unknown and current therapeutic approaches are symptomatic only, and frequently inconvenient or invasive. Muscular contribution to AIS is widely recognized, although it did not translate to clinical routine as yet. Muscle asymmetry has been documented by pathological examinations as well as systemic muscle disorders frequently leading to scoliosis. It has been also reported numerous genetic, metabolic and radiological alterations in patients with AIS, which are linked to muscular and neuromuscular aspects. Therefore, muscles might be considered an attractive and still insufficiently exploited therapeutic target for AIS. Stem cell-based regenerative medicine is rapidly gaining momentum based on the tremendous progress in understanding of developmental biology. It comes also with a toolbox of various stem cells such as satellite cells or mesenchymal stem cells, which could be transplanted; also, the knowledge acquired in research on regenerative medicine can be applied to manipulation of endogenous stem cells to obtain desired therapeutic goals. Importantly, paravertebral muscles are located relatively superficially; therefore, they can be an easy target for minimally invasive approaches to treatment of AIS. It comes in pair with a fast progress in image guidance, which allows for precise delivery of therapeutic agents, including stem cells to various organs such as brain, muscles, and others. Summing up, it seems that there is a link between AIS, muscles, and stem cells, which might be worth of further investigations with a long-term goal of setting foundations for eventual bench-to-bedside translation.
Collapse
Affiliation(s)
- Edyta Brzoska
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Lukasz Kalkowski
- 2Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Kamil Kowalski
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Pawel Michalski
- 3Spine Surgery Department, Institute of Mother and Child, Warsaw, Poland
| | - Pawel Kowalczyk
- 4Department of Neurosurgery, Children's Memorial Health Institute, Warsaw, Poland
| | - Bartosz Mierzejewski
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Piotr Walczak
- 5Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,6Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Maria A Ciemerych
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Miroslaw Janowski
- 5Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,6Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
18
|
Mechanisms linking adipose tissue inflammation to cardiac hypertrophy and fibrosis. Clin Sci (Lond) 2020; 133:2329-2344. [PMID: 31777927 DOI: 10.1042/cs20190578] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/08/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022]
Abstract
Adipose tissue is classically recognized as the primary site of lipid storage, but in recent years has garnered appreciation for its broad role as an endocrine organ comprising multiple cell types whose collective secretome, termed as adipokines, is highly interdependent on metabolic homeostasis and inflammatory state. Anatomical location (e.g. visceral, subcutaneous, epicardial etc) and cellular composition of adipose tissue (e.g. white, beige, and brown adipocytes, macrophages etc.) also plays a critical role in determining its response to metabolic state, the resulting secretome, and its potential impact on remote tissues. Compared with other tissues, the heart has an extremely high and constant demand for energy generation, of which most is derived from oxidation of fatty acids. Availability of this fatty acid fuel source is dependent on adipose tissue, but evidence is mounting that adipose tissue plays a much broader role in cardiovascular physiology. In this review, we discuss the impact of the brown, subcutaneous, and visceral white, perivascular (PVAT), and epicardial adipose tissue (EAT) secretome on the development and progression of cardiovascular disease (CVD), with a particular focus on cardiac hypertrophy and fibrosis.
Collapse
|
19
|
Kim HW, Belin de Chantemèle EJ, Weintraub NL. Perivascular Adipocytes in Vascular Disease. Arterioscler Thromb Vasc Biol 2019; 39:2220-2227. [PMID: 31510794 DOI: 10.1161/atvbaha.119.312304] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Perivascular adipocytes residing in the vascular adventitia are recognized as distinct endocrine cells capable of responding to inflammatory stimuli and communicating with the sympathetic nervous system and adjacent blood vessel cells, thereby releasing adipocytokines and other signaling mediators to maintain vascular homeostasis. Perivascular adipocytes exhibit phenotypic heterogeneity (both white and brown adipocytes) and become dysfunctional in conditions, such as diet-induced obesity, thus promoting vascular inflammation, vasoconstriction, and smooth muscle cell proliferation to potentially contribute to the development of vascular diseases, such as atherosclerosis, hypertension, and aortic aneurysms. Although accumulating data have advanced our understanding of the role of perivascular adipocytes in modulating vascular function, their impact on vascular disease, particularly in humans, remains to be fully defined. This brief review will discuss the mechanisms whereby perivascular adipocytes regulate vascular disease, with a particular emphasis on recent findings and current limitations in the field of research.
Collapse
Affiliation(s)
- Ha Won Kim
- From the Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University
| | - Eric J Belin de Chantemèle
- From the Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University
| | - Neal L Weintraub
- From the Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University
| |
Collapse
|
20
|
Zhang L, Wang Y, Zhou N, Feng Y, Yang X. Cyclic tensile stress promotes osteogenic differentiation of adipose stem cells via ERK and p38 pathways. Stem Cell Res 2019; 37:101433. [PMID: 31005788 DOI: 10.1016/j.scr.2019.101433] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/03/2019] [Accepted: 04/06/2019] [Indexed: 02/05/2023] Open
Abstract
The present study aimed to elucidate whether extracellular signal-regulated kinases 1/2 (ERK1/2) and p38 mitogen-activated protein kinases pathways participate in the transduction of mechanical stretch exerted on adipose stem cells (ASCs) into intracellular osteogenic signals, and if so whether both pathways have time-dependent feature. Rat ASCs were cultured in osteogenic medium for 72 h and assigned into three sets, namely ERK1/2 inhibitor treated set, p38 inhibitor treated set, and the control set. After inhibitor treatment, all cells were subjected to cyclic stretch(2000 με, 1 Hz) on a four-point bending mechanical loading device. Protein and mRNA samples were acquired at six time points: 0, 15 min, 30 min, 1 h, 2 h and 6 h. Western blot showed phosphorylation level of ERK1/2 was elevated by cyclic tensile stress at all time points, while p38 at 15 min, 30 min and 1 h, and the elevation can be completely blocked by corresponding inhibitors. The treatment by ERK1/2 inhibitor was shown to antagonize the up-regulation of osteogenic genes bone morphogenetic protein 2 (BMP-2) and runt-related transcription factor 2 (Runx2) by mechanical stretch at 15 min and 6 h, whereas p38 inhibitor took effect at 15 min only. The results suggested both ERK and p38 could be positive mediators of stretch-induced osteogenic differentiation of ASCs, and ERK stimulate the stretch-induced osteogenic differentiation at both early and late stages while p38 responds to mechanical stretch in a more rapid fashion.
Collapse
Affiliation(s)
- Liang Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yingkai Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Nan Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yuzhang Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xingmei Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
21
|
Takahashi H, Sakata N, Yoshimatsu G, Hasegawa S, Kodama S. Regenerative and Transplantation Medicine: Cellular Therapy Using Adipose Tissue-Derived Mesenchymal Stromal Cells for Type 1 Diabetes Mellitus. J Clin Med 2019; 8:249. [PMID: 30781427 PMCID: PMC6406504 DOI: 10.3390/jcm8020249] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/09/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is caused by the autoimmune targeting of pancreatic β-cells, and, in the advanced stage, severe hypoinsulinemia due to islet destruction. In patients with T1DM, continuous exogenous insulin therapy cannot be avoided. However, an insufficient dose of insulin easily induces extreme hyperglycemia or diabetic ketoacidosis, and intensive insulin therapy may cause hypoglycemic symptoms including hypoglycemic shock. While these insulin therapies are efficacious in most patients, some additional therapies are warranted to support the control of blood glucose levels and reduce the risk of hypoglycemia in patients who respond poorly despite receiving appropriate treatment. There has been a recent gain in the popularity of cellular therapies using mesenchymal stromal cells (MSCs) in various clinical fields, owing to their multipotentiality, capacity for self-renewal, and regenerative and immunomodulatory potential. In particular, adipose tissue-derived MSCs (ADMSCs) have become a focus in the clinical setting due to the abundance and easy isolation of these cells. In this review, we outline the possible therapeutic benefits of ADMSC for the treatment of T1DM.
Collapse
Affiliation(s)
- Hiroyuki Takahashi
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Naoaki Sakata
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Gumpei Yoshimatsu
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Suguru Hasegawa
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Shohta Kodama
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| |
Collapse
|
22
|
Abstract
Adequate bone remodeling may be a primary parameter for long-term successful complication-free dental implant treatment. A 1.8-mm osseous thickness around dental implants is thought to be the minimum thickness for adequate vasculature for osteocyte nutrition and function. A dental implant does not provide progenitor cells or angiogenic or osteogenic factors. Thus, the surrounding bone may need to have a 1.8-mm thickness to accommodate the vasculature necessary for nutrients for appropriate remodeling. Additionally, the 1.8-mm dimension may provide for mechanical load resistance. There is no evidence to illustrate the physiologic need for the 1.8-mm dimension. This dimension requirement is based on clinical outcome observations. Basic science research for bone survival around dental implants is needed.
Collapse
|
23
|
Rojas-Rodriguez R, Lujan-Hernandez J, Min SY, DeSouza T, Teebagy P, Desai A, Tessier H, Slamin R, Siegel-Reamer L, Berg C, Baez A, Lalikos J, Corvera S. Generation of Functional Human Adipose Tissue in Mice from Primed Progenitor Cells. Tissue Eng Part A 2019; 25:842-854. [PMID: 30306830 PMCID: PMC6590775 DOI: 10.1089/ten.tea.2018.0067] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Adipose tissue (AT) is used extensively in reconstructive and regenerative therapies, but transplanted fat often undergoes cell death, leading to inflammation, calcification, and requirement for further revision surgery. Previously, we have found that mesenchymal progenitor cells within human AT can proliferate in three-dimensional culture under proangiogenic conditions. These cells (primed ADipose progenitor cells, PADS) robustly differentiate into adipocytes in vitro (ad-PADS). The goal of this study is to determine whether ad-PADS can form structured AT in vivo, with potential for use in surgical applications. Grafts formed from ad-PADS were compared to grafts formed from AT obtained by liposuction after implantation into nude mice. Graft volume was measured by microcomputed tomography scanning, and the functionality of cells within the graft was assessed by quantifying circulating human adiponectin. The degree of graft vascularization by donor or host vessels and the content of human or mouse adipocytes within the graft were measured using species-specific endothelial and adipocyte-specific quantitative real time polymerase chain reaction probes, and histochemistry with mouse and human-specific lectins. Our results show that ad-PADS grafted subcutaneously into nude mice induce robust vascularization from the host, continue to increase in volume over time, express the human adipocyte marker PLIN1 at levels comparable to human AT, and secrete increasing amounts of human adiponectin into the mouse circulation. In contrast, grafts composed of AT fragments obtained by liposuction become less vascularized, develop regions of calcification and decreased content of PLIN1, and secrete lower amounts of adiponectin per unit volume. Enrichment of liposuction tissue with ad-PADS improves vascularization, indicating that ad-PADS may be proangiogenic. Mechanistically, ad-PADS express an extracellular matrix gene signature that includes elements previously associated with small vessel development (COL4A1). Thus, through the formation of a proangiogenic environment, ad-PADS can form functional AT with capacity for long-term survival, and can potentially be used to improve outcomes in reconstructive and regenerative medicine.
Collapse
Affiliation(s)
- Raziel Rojas-Rodriguez
- 1 Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jorge Lujan-Hernandez
- 2 Department of Surgery, University of Massachusetts Medical School and UMASS Memorial Medical Center, Worcester, Massachusetts
| | - So Yun Min
- 1 Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Tiffany DeSouza
- 1 Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Patrick Teebagy
- 2 Department of Surgery, University of Massachusetts Medical School and UMASS Memorial Medical Center, Worcester, Massachusetts
| | - Anand Desai
- 1 Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Heather Tessier
- 2 Department of Surgery, University of Massachusetts Medical School and UMASS Memorial Medical Center, Worcester, Massachusetts
| | - Robert Slamin
- 2 Department of Surgery, University of Massachusetts Medical School and UMASS Memorial Medical Center, Worcester, Massachusetts
| | - Leah Siegel-Reamer
- 2 Department of Surgery, University of Massachusetts Medical School and UMASS Memorial Medical Center, Worcester, Massachusetts
| | - Cara Berg
- 1 Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Angel Baez
- 2 Department of Surgery, University of Massachusetts Medical School and UMASS Memorial Medical Center, Worcester, Massachusetts
| | - Janice Lalikos
- 2 Department of Surgery, University of Massachusetts Medical School and UMASS Memorial Medical Center, Worcester, Massachusetts
| | - Silvia Corvera
- 1 Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
24
|
Jin YZ, Lee JH. Mesenchymal Stem Cell Therapy for Bone Regeneration. Clin Orthop Surg 2018; 10:271-278. [PMID: 30174801 PMCID: PMC6107811 DOI: 10.4055/cios.2018.10.3.271] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/27/2018] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been used in clinic for approximately 20 years. During this period, various new populations of MSCs have been found or manipulated. However, their characters and relative strength for bone regeneration have not been well known. For a comprehensive understanding of MSCs, we reviewed the literature on the multipotent cells ranging from the definition to the current research progress for bone regeneration. Based on our literature review, bone marrow MSCs have been most widely studied and utilized in clinical settings. Among other populations of MSCs, adipose-derived MSCs and perivascular MSCs might be potential candidates for bone regeneration, whose efficacy and safety still require further investigation.
Collapse
Affiliation(s)
- Yuan-Zhe Jin
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Hyup Lee
- Department of Orthopedic Surgery, Seoul National University College of Medicine, Seoul, Korea.,Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul, Korea.,Institute of Medical and Biological Engineering, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
25
|
Periasamy R, Elshaer SL, Gangaraju R. CD140b (PDGFRβ) signaling in adipose-derived stem cells mediates angiogenic behavior of retinal endothelial cells. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018; 5:1-9. [PMID: 30976657 DOI: 10.1007/s40883-018-0068-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Adipose-derived stem cells (ASCs) are multipotent mesenchymal progenitor cells that have functional and phenotypic overlap with pericytes lining microvessels in adipose tissue. The role of CD140b [platelet-derived growth factor receptor- β (PDGFR-β)], a constitutive marker expressed by ASCs, in the angiogenic behavior of human retinal endothelial cells (HREs) is not known. CD140b was knocked down in ASCs using targeted siRNA and lipofectamine transfection protocol. Both CD140b+ and CD140b- ASCs were tested for their proliferation (WST-1 reagent), adhesion (laminin-1 coated plates), and migration (wound-scratch assay). Angiogenic effect of CD140b+ and CD140b- ASCs on HREs was examined by co-culturing ASCs:HREs in 12:1 ratio for 6 days followed by visualization of vascular network by Isolectin B4 staining. The RayBio® Membrane-Based Antibody Array was used to assess differences in human cytokines released by CD140b+ or CD140b- ASCs. Knockdown of CD140b in ASCs resulted in a significant 50% decrease in proliferation rate, 25% decrease in adhesion ability to Laminin-1, and 50% decrease in migration rate, as compared to CD140b+ ASCs. Direct contact of ASCs expressing CD140b+ with HREs resulted in robust vascular network formation that was significantly reduced with using CD140b- ASCs. Of the 80 proteins tested, 45 proteins remained unchanged (>0.5-<1.5 fold), 6 proteins including IL-10 downregulated (<0.5 fold) and 29 proteins including IL-16 & TNF-β were upregulated (>1.5 fold) in CD140b- ASCs compared to CD140b+ ASCs. Our data demonstrate a substantial role for CD140b in the intrinsic abilities of ASCs and their angiogenic influence on HREs. Future studies are needed to fully explore the signaling of CD140b in ASCs in vivo for retinal regeneration.
Collapse
Affiliation(s)
- Ramesh Periasamy
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN, 38163. USA
| | - Sally L Elshaer
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN, 38163. USA
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Sciences Center, Memphis, TN, 38163. USA.,Anatomy and neurobiology, University of Tennessee Health Sciences Center, Memphis, TN, 38163. USA
| |
Collapse
|
26
|
Schwalie PC, Dong H, Zachara M, Russeil J, Alpern D, Akchiche N, Caprara C, Sun W, Schlaudraff KU, Soldati G, Wolfrum C, Deplancke B. A stromal cell population that inhibits adipogenesis in mammalian fat depots. Nature 2018; 559:103-108. [PMID: 29925944 DOI: 10.1038/s41586-018-0226-8] [Citation(s) in RCA: 321] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/16/2018] [Indexed: 02/07/2023]
Abstract
Adipocyte development and differentiation have an important role in the aetiology of obesity and its co-morbidities1,2. Although multiple studies have investigated the adipogenic stem and precursor cells that give rise to mature adipocytes3-14, our understanding of their in vivo origin and properties is incomplete2,15,16. This is partially due to the highly heterogeneous and unstructured nature of adipose tissue depots17, which has proven difficult to molecularly dissect using classical approaches such as fluorescence-activated cell sorting and Cre-lox lines based on candidate marker genes16,18. Here, using the resolving power of single-cell transcriptomics19 in a mouse model, we reveal distinct subpopulations of adipose stem and precursor cells in the stromal vascular fraction of subcutaneous adipose tissue. We identify one of these subpopulations as CD142+ adipogenesis-regulatory cells, which can suppress adipocyte formation in vivo and in vitro in a paracrine manner. We show that adipogenesis-regulatory cells are refractory to adipogenesis and that they are functionally conserved in humans. Our findings point to a potentially critical role for adipogenesis-regulatory cells in modulating adipose tissue plasticity, which is linked to metabolic control, differential insulin sensitivity and type 2 diabetes.
Collapse
Affiliation(s)
- Petra C Schwalie
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Hua Dong
- Institute of Food Nutrition and Health, Eidgenössische Technische Hochschule Zürich, Schwerzenbach, Switzerland
| | - Magda Zachara
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Julie Russeil
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Daniel Alpern
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Nassila Akchiche
- Institute of Food Nutrition and Health, Eidgenössische Technische Hochschule Zürich, Schwerzenbach, Switzerland
| | | | - Wenfei Sun
- Institute of Food Nutrition and Health, Eidgenössische Technische Hochschule Zürich, Schwerzenbach, Switzerland
| | | | | | - Christian Wolfrum
- Institute of Food Nutrition and Health, Eidgenössische Technische Hochschule Zürich, Schwerzenbach, Switzerland.
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL) and Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
27
|
Schoettl T, Fischer IP, Ussar S. Heterogeneity of adipose tissue in development and metabolic function. ACTA ACUST UNITED AC 2018. [PMID: 29514879 DOI: 10.1242/jeb.162958] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adipose tissue is a central metabolic organ. Unlike other organs, adipose tissue is compartmentalized into individual depots and distributed throughout the body. These different adipose depots show major functional differences and risk associations for developing metabolic syndrome. Recent advances in lineage tracing demonstrate that individual adipose depots are composed of adipocytes that are derived from distinct precursor populations, giving rise to different populations of energy-storing white adipocytes. Moreover, distinct lineages of energy-dissipating brown and beige adipocytes exist in discrete depots or within white adipose tissue depots. In this Review, we discuss developmental and functional heterogeneity, as well as sexual dimorphism, between and within individual adipose tissue depots. We highlight current data relating to the differences between subcutaneous and visceral white adipose tissue in the development of metabolic dysfunction, with special emphasis on adipose tissue expansion and remodeling of the extracellular matrix. Moreover, we provide a detailed overview of adipose tissue development as well as the consensus and controversies relating to adult adipocyte precursor populations.
Collapse
Affiliation(s)
- Theresa Schoettl
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Germany.,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Ingrid P Fischer
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Germany.,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.,Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333 Munich, Germany
| | - Siegfried Ussar
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Germany .,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| |
Collapse
|
28
|
Angiogenesis in a 3D model containing adipose tissue stem cells and endothelial cells is mediated by canonical Wnt signaling. Bone Res 2017; 5:17048. [PMID: 29263938 PMCID: PMC5727463 DOI: 10.1038/boneres.2017.48] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 03/25/2017] [Accepted: 04/10/2017] [Indexed: 02/05/2023] Open
Abstract
Adipose-derived stromal cells (ASCs) have gained great attention in regenerative medicine. Progress in our understanding of adult neovascularization further suggests the potential of ASCs in promoting vascular regeneration, although the specific cues that stimulate their angiogenic behavior remain controversial. In this study, we established a three-dimensional (3D) angiogenesis model by co-culturing ASCs and endothelial cells (ECs) in collagen gel and found that ASC-EC-instructed angiogenesis was regulated by the canonical Wnt pathway. Furthermore, the angiogenesis that occurred in implants collected after injections of our collagen gel-based 3D angiogenesis model into nude mice was confirmed to be functional and also regulated by the canonical Wnt pathway. Wnt regulation of angiogenesis involving changes in vessel length, vessel density, vessel sprout, and connection numbers occurred in our system. Wnt signaling was then shown to regulate ASC-mediated paracrine signaling during angiogenesis through the nuclear translocation of β-catenin after its cytoplasmic accumulation in both ASCs and ECs. This translocation enhanced the expression of nuclear co-factor Lef-1 and cyclin D1 and activated the angiogenic transcription of vascular endothelial growth factor A (VEGFA), basic fibroblast growth factor (bFGF), and insulin-like growth factor 1 (IGF-1). The angiogenesis process in the 3D collagen model appeared to follow canonical Wnt signaling, and this model can help us understand the importance of the canonical Wnt pathway in the use of ASCs in vascular regeneration.
Collapse
|
29
|
Horimatsu T, Kim HW, Weintraub NL. The Role of Perivascular Adipose Tissue in Non-atherosclerotic Vascular Disease. Front Physiol 2017; 8:969. [PMID: 29234289 PMCID: PMC5712360 DOI: 10.3389/fphys.2017.00969] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/14/2017] [Indexed: 12/30/2022] Open
Abstract
Perivascular adipose tissue (PVAT) surrounds most large blood vessels and plays an important role in vascular homeostasis. PVAT releases various chemokines and adipocytokines, functioning in an endocrine and paracrine manner to regulate vascular signaling and inflammation. Mounting evidence suggests that PVAT plays an important role in atherosclerosis and hypertension; however, the role of PVAT in non-atherosclerotic vascular diseases, including neointimal formation, aortic aneurysm, arterial stiffness and vasculitis, has received far less attention. Increasing evidence suggests that PVAT responds to mechanical endovascular injury and regulates the subsequent formation of neointima via factors that promote smooth muscle cell growth, adventitial inflammation and neovascularization. Circumstantial evidence also links PVAT to the pathogenesis of aortic aneurysms and vasculitic syndromes, such as Takayasu's arteritis, where infiltration and migration of inflammatory cells from PVAT into the vascular wall may play a contributory role. Moreover, in obesity, PVAT has been implicated to promote stiffness of elastic arteries via the production of reactive oxygen species. This review will discuss the growing body of data and mechanisms linking PVAT to the pathogenesis of non-atherosclerotic vascular diseases in experimental animal models and in humans.
Collapse
Affiliation(s)
- Tetsuo Horimatsu
- Division of Cardiology, Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Ha Won Kim
- Division of Cardiology, Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Neal L Weintraub
- Division of Cardiology, Department of Medicine, Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
30
|
Scioli MG, Bielli A, Gentile P, Cervelli V, Orlandi A. Combined treatment with platelet-rich plasma and insulin favours chondrogenic and osteogenic differentiation of human adipose-derived stem cells in three-dimensional collagen scaffolds. J Tissue Eng Regen Med 2017; 11:2398-2410. [PMID: 27074878 DOI: 10.1002/term.2139] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 11/18/2015] [Accepted: 12/10/2015] [Indexed: 02/06/2023]
Abstract
Osteochondral lesions due to injury or other pathology commonly result in the development of osteoarthritis and progressive joint destruction. Bioengineered scaffolds are widely studied for regenerative surgery strategies in osteochondral defect management, also combining the use of stem cells, growth factors and hormones. The utility in tissue engineering of human adipose-derived stem cells (ASCs) isolated from adipose tissue has been widely noted. Autologous platelet-rich plasma (PRP) represents an alternative strategy in regenerative medicine for the local release of endogenous growth factors and hormones. Here we compared the effects of three-dimensional (3D) collagen type I scaffold culture and combined treatment with PRP and human recombinant insulin on the chondro-/osteogenic differentiation of ASCs. Histochemical and biomolecular analyses demonstrated that chondro-/osteogenic differentiation was increased in ASC-populated 3D collagen scaffolds compared with two-dimensional (2D) plastic dish culture. Chondro-/osteogenic differentiation was further enhanced in the presence of combined PRP (5% v/v) and insulin (100 nm) treatment. In addition, chondro-/osteogenic differentiation associated with the contraction of ASC-populated 3D collagen scaffold and increased β1/β3-integrin expression. Inhibition studies demonstrated that PRP/insulin-induced chondro-/osteogenic differentiation is independent of insulin-like growth factor 1 receptor (IGF-1R) and mammalian target of rapamycin (mTOR) signalling; IGF-R1/mTOR inhibition even enhanced ASC chondro-/osteogenic differentiation. Our findings underline that 3D collagen scaffold culture in association with platelet-derived growth factors and insulin favour the chondro-/osteogenic differentiation of ASCs, suggesting new translational applications in regenerative medicine for the management of osteochondral defects. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Maria Giovanna Scioli
- Institute of Anatomical Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Alessandra Bielli
- Institute of Anatomical Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Pietro Gentile
- Plastic and Reconstructive Surgery, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Valerio Cervelli
- Plastic and Reconstructive Surgery, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Augusto Orlandi
- Institute of Anatomical Pathology, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| |
Collapse
|
31
|
Application of adipose-derived stromal cells in fat grafting: Basic science and literature review. Exp Ther Med 2017; 14:2415-2423. [PMID: 28962175 PMCID: PMC5609216 DOI: 10.3892/etm.2017.4811] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/06/2017] [Indexed: 02/07/2023] Open
Abstract
Autologous fat is considered the ideal material for soft-tissue augmentation in plastic and reconstructive surgery. The primary drawback of autologous fat grafting is the high resorption rate. The isolation of mesenchymal stem cells from adipose tissue inevitably led to research focusing on the study of combined transplantation of autologous fat and adipose derived stem cells (ADSCs) and introduced the theory of ‘cell-assisted lipotransfer’. Transplantation of ADSCs is a promising strategy, due to the high proliferative capacity of stem cells, their potential to induce paracrine signalling and ability to differentiate into adipocytes and vascular cells. The current study examined the literature for clinical and experimental studies on cell-assisted lipotransfer to assess the efficacy of this novel technique when compared with traditional fat grafting. A total of 30 studies were included in the present review. The current study demonstrates that cell-assisted lipotransfer has improved efficacy compared with conventional fat grafting. Despite relatively positive outcomes, further investigation is required to establish a consensus in cell-assisted lipotransfer.
Collapse
|
32
|
Jiang Y, Berry DC, Jo A, Tang W, Arpke RW, Kyba M, Graff JM. A PPARγ transcriptional cascade directs adipose progenitor cell-niche interaction and niche expansion. Nat Commun 2017. [PMID: 28649987 PMCID: PMC5490270 DOI: 10.1038/ncomms15926] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Adipose progenitor cells (APCs) reside in a vascular niche, located within the perivascular compartment of adipose tissue blood vessels. Yet, the signals and mechanisms that govern adipose vascular niche formation and APC niche interaction are unknown. Here we show that the assembly and maintenance of the adipose vascular niche is controlled by PPARγ acting within APCs. PPARγ triggers a molecular hierarchy that induces vascular sprouting, APC vessel niche affinity and APC vessel occupancy. Mechanistically, PPARγ transcriptionally activates PDGFRβ and VEGF. APC expression and activation of PDGFRβ promotes the recruitment and retention of APCs to the niche. Pharmacologically, targeting PDGFRβ disrupts APC niche contact thus blocking adipose tissue expansion. Moreover, enhanced APC expression of VEGF stimulates endothelial cell proliferation and expands the adipose niche. Consequently, APC niche communication and retention are boosted by VEGF thereby impairing adipogenesis. Our data indicate that APCs direct adipose tissue niche expansion via a PPARγ-initiated PDGFRβ and VEGF transcriptional axis. Adipocyte progenitor cells (APCs) are found tethered to adipose tissue blood vessel walls and can differentiate into adipocytes. Here the authors show that PPARγ controls angiogenesis by stimulating APC–blood vessel interaction and retention via a transcriptional network that includes PDGFRβ and VEGF.
Collapse
Affiliation(s)
- Yuwei Jiang
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Daniel C Berry
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Ayoung Jo
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Wei Tang
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Robert W Arpke
- Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Michael Kyba
- Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA.,Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Jonathan M Graff
- Division of Endocrinology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
33
|
Priglinger E, Wurzer C, Steffenhagen C, Maier J, Hofer V, Peterbauer A, Nuernberger S, Redl H, Wolbank S, Sandhofer M. The adipose tissue-derived stromal vascular fraction cells from lipedema patients: Are they different? Cytotherapy 2017; 19:849-860. [PMID: 28454682 DOI: 10.1016/j.jcyt.2017.03.073] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 01/18/2023]
Abstract
BACKGROUND AIMS Lipedema is a hormone-related disease of women characterized by enlargement of the extremities caused by subcutaneous deposition of adipose tissue. In healthy patients application of autologous adipose tissue-derived cells has shown great potential in several clinical studies for engrafting of soft tissue reconstruction in recent decades. The majority of these studies have used the stromal vascular fraction (SVF), a heterogeneous cell population containing adipose-derived stromal/stem cells (ASC), among others. Because cell identity and regenerative properties might be affected by the health condition of patients, we characterized the SVF cells of 30 lipedema patients in comparison to 22 healthy patients. METHODS SVF cells were analyzed regarding cell yield, viability, adenosine triphosphate content, colony forming units and proliferative capacity, as well as surface marker profile and differentiation potential in vitro. RESULTS Our results demonstrated a significantly enhanced SVF cell yield isolated from lipedema compared with healthy patients. In contrast, the adipogenic differentiation potential of SVF cells isolated from lipedema patients was significantly reduced compared with healthy patients. Interestingly, expression of the mesenchymal marker CD90 and the endothelial/pericytic marker CD146 was significantly enhanced when isolated from lipedema patients. DISCUSSION The enhanced number of CD90+ and CD146+ cells could explain the increased cell yield because the other tested surface marker were not reduced in lipedema patients. Because the cellular mechanism and composition in lipedema is largely unknown, our findings might contribute to a better understanding of its etiology.
Collapse
Affiliation(s)
- Eleni Priglinger
- AUVA Research Center, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Linz, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| | - Christoph Wurzer
- AUVA Research Center, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Linz, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria; Liporegena GmbH, Breitenfurt, Austria
| | - Carolin Steffenhagen
- AUVA Research Center, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Linz, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Julia Maier
- AUVA Research Center, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Linz, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Victoria Hofer
- Faculty of Medicine/Dental Medicine, Danube Private University, Krems-Stein, Austria; Austrian Academy of Cosmetic Surgery and Aesthetic Medicine, Linz, Austria
| | - Anja Peterbauer
- Austrian Cluster for Tissue Regeneration, Vienna, Austria; Red Cross Blood Transfusion Service of Upper Austria, Linz, Austria
| | - Sylvia Nuernberger
- Austrian Cluster for Tissue Regeneration, Vienna, Austria; Bernhard Gottlieb University Clinic of Dentistry, Universitätsklinik für Zahn-, Mund- und Kieferheilkunde Ges.m.b.H, Vienna, Austria; Department of Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Heinz Redl
- AUVA Research Center, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Linz, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Susanne Wolbank
- AUVA Research Center, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Linz, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Matthias Sandhofer
- Austrian Academy of Cosmetic Surgery and Aesthetic Medicine, Linz, Austria
| |
Collapse
|
34
|
Gennai A, Zambelli A, Repaci E, Quarto R, Baldelli I, Fraternali G, Bernardini FP. Skin Rejuvenation and Volume Enhancement with the Micro Superficial Enhanced Fluid Fat Injection (M-SEFFI) for Skin Aging of the Periocular and Perioral Regions. Aesthet Surg J 2017; 37:14-23. [PMID: 27241362 DOI: 10.1093/asj/sjw084] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Adipose-derived stromal and stem cells (ADSC) in autologous fat promises regenerative advantages, and injected into the dermal and subdermal layers, enhances rejuvenation and volume. However, extremely superficial fat injection with current techniques is limited. OBJECTIVES Efficacy and viability evaluation of fat harvested with extremely small side port (0.3 mm) cannulae without further tissue manipulation for the correction of aging/thin skin in the periocular and perioral regions. METHODS Micro-superficial enhanced fluid fat injection (M-SEFFI) harvests adipose tissue with a multi-perforated cannula (0.3 mm), and autologous platelet rich plasma (PRP) is added. The tissue is injected into the dermal region of the periocular and perioral zones. Efficacy and viability were evaluated by histological and cell culture analysis. Clinical assessment included retrospective evaluation according to 1 = no effect, 2 = fair effect, 3 = good effect, 4 = excellent effect. RESULTS Between June 2014 and July 2015, 65 patients (7 men; mean age 49.7 years) were treated with M-SEFFI. No intraoperative complications or visible lumpiness were recorded. Analysis demonstrated mature, viable adipocytes with a strong stromal component. Following PRP addition, there was a greater proliferation noted in the M-SEFFI compared to the SEFFI (0.5 mm). Mean follow-up was 4.1 months. Clinical assessment by surgeons and patients at 1 month was 3.52 and 3.74, and 6 months 3.06 and 2.6 respectively. CONCLUSIONS M-SEFFI is effective and viable for lump free skin rejuvenation and volume enhancement, through the extraction of smoother ADSC rich, autologous fat tissue that does not require further tissue manipulation, to correct skin aging. LEVEL OF EVIDENCE 4 Therapeutic.
Collapse
Affiliation(s)
- Alessandro Gennai
- Dr Gennai is a plastic surgeon in private practice in Bologna and Milan, Italy. Drs Zambelli and Bernardini are oculoplastic surgeons in private practice in Genova, Italy. Dr Repaci is a Researcher and Dr Quarto is the Chairman, Department of Experimental Medicine, University of Genova, Genova, Italy. Dr Baldelli is a Plastic Surgeon, Department of Integrated Surgical and Diagnostic Sciences, University of Genova, Genova, Italy. Dr Fraternali is a Pathologist, Department of Pathology, University of Genova, Genova, Italy
| | - Alessandra Zambelli
- Dr Gennai is a plastic surgeon in private practice in Bologna and Milan, Italy. Drs Zambelli and Bernardini are oculoplastic surgeons in private practice in Genova, Italy. Dr Repaci is a Researcher and Dr Quarto is the Chairman, Department of Experimental Medicine, University of Genova, Genova, Italy. Dr Baldelli is a Plastic Surgeon, Department of Integrated Surgical and Diagnostic Sciences, University of Genova, Genova, Italy. Dr Fraternali is a Pathologist, Department of Pathology, University of Genova, Genova, Italy
| | - Erica Repaci
- Dr Gennai is a plastic surgeon in private practice in Bologna and Milan, Italy. Drs Zambelli and Bernardini are oculoplastic surgeons in private practice in Genova, Italy. Dr Repaci is a Researcher and Dr Quarto is the Chairman, Department of Experimental Medicine, University of Genova, Genova, Italy. Dr Baldelli is a Plastic Surgeon, Department of Integrated Surgical and Diagnostic Sciences, University of Genova, Genova, Italy. Dr Fraternali is a Pathologist, Department of Pathology, University of Genova, Genova, Italy
| | - Rodolfo Quarto
- Dr Gennai is a plastic surgeon in private practice in Bologna and Milan, Italy. Drs Zambelli and Bernardini are oculoplastic surgeons in private practice in Genova, Italy. Dr Repaci is a Researcher and Dr Quarto is the Chairman, Department of Experimental Medicine, University of Genova, Genova, Italy. Dr Baldelli is a Plastic Surgeon, Department of Integrated Surgical and Diagnostic Sciences, University of Genova, Genova, Italy. Dr Fraternali is a Pathologist, Department of Pathology, University of Genova, Genova, Italy
| | - Ilaria Baldelli
- Dr Gennai is a plastic surgeon in private practice in Bologna and Milan, Italy. Drs Zambelli and Bernardini are oculoplastic surgeons in private practice in Genova, Italy. Dr Repaci is a Researcher and Dr Quarto is the Chairman, Department of Experimental Medicine, University of Genova, Genova, Italy. Dr Baldelli is a Plastic Surgeon, Department of Integrated Surgical and Diagnostic Sciences, University of Genova, Genova, Italy. Dr Fraternali is a Pathologist, Department of Pathology, University of Genova, Genova, Italy
| | - Giulio Fraternali
- Dr Gennai is a plastic surgeon in private practice in Bologna and Milan, Italy. Drs Zambelli and Bernardini are oculoplastic surgeons in private practice in Genova, Italy. Dr Repaci is a Researcher and Dr Quarto is the Chairman, Department of Experimental Medicine, University of Genova, Genova, Italy. Dr Baldelli is a Plastic Surgeon, Department of Integrated Surgical and Diagnostic Sciences, University of Genova, Genova, Italy. Dr Fraternali is a Pathologist, Department of Pathology, University of Genova, Genova, Italy
| | - Francesco P Bernardini
- Dr Gennai is a plastic surgeon in private practice in Bologna and Milan, Italy. Drs Zambelli and Bernardini are oculoplastic surgeons in private practice in Genova, Italy. Dr Repaci is a Researcher and Dr Quarto is the Chairman, Department of Experimental Medicine, University of Genova, Genova, Italy. Dr Baldelli is a Plastic Surgeon, Department of Integrated Surgical and Diagnostic Sciences, University of Genova, Genova, Italy. Dr Fraternali is a Pathologist, Department of Pathology, University of Genova, Genova, Italy
| |
Collapse
|
35
|
Youngstrom DW, LaDow JE, Barrett JG. Tenogenesis of bone marrow-, adipose-, and tendon-derived stem cells in a dynamic bioreactor. Connect Tissue Res 2016; 57:454-465. [PMID: 27028488 DOI: 10.3109/03008207.2015.1117458] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Tendons are frequently damaged and fail to regenerate, leading to pain, loss of function, and reduced quality of life. Mesenchymal stem cells (MSCs) possess clinically useful tissue-regenerative properties and have been exploited for use in tendon tissue engineering and cell therapy. However, MSCs exhibit phenotypic heterogeneity based on the donor tissue used, and the efficacy of cell-based treatment modalities may be improved by optimizing cell source based on relative differentiation capacity. Equine MSCs were isolated from bone marrow (BM), adipose (AD), and tendon (TN), expanded in monolayer prior to seeding on decellularized tendon scaffolds (DTS), and cell-laden constructs were placed in a bioreactor designed to mimic the biophysical environment of the tendon. It was hypothesized that TN MSCs would differentiate toward a tendon cell phenotype better than BM and AD MSCs in response to a conditioning period involving cyclic mechanical stimulation for 1 hour per day at 3% strain and 0.33 Hz. All cell types integrated into DTS adopted an elongated morphology similar to tenocytes, expressed tendon marker genes, and improved tissue mechanical properties after 11 days. TN MSCs expressed the greatest levels of scleraxis, collagen type-I, and cartilage oligomeric matrix protein. Major histocompatibility class-II protein mRNA expression was not detected in any of the MSC types, suggesting low immunogenicity for allogeneic transplantation. The results suggest that TN MSCs are the ideal cell type for regenerative medicine therapies for tendinopathies, exhibiting the most mature tendon-like phenotype in vitro. When TN MSCs are unavailable, BM or AD MSCs may serve as robust alternatives.
Collapse
Affiliation(s)
- Daniel W Youngstrom
- a Program in Biomedical and Veterinary Sciences, Marion duPont Scott Equine Medical Center , Virginia Tech , Leesburg , VA , USA
| | - Jade E LaDow
- a Program in Biomedical and Veterinary Sciences, Marion duPont Scott Equine Medical Center , Virginia Tech , Leesburg , VA , USA
| | - Jennifer G Barrett
- b Department of Large Animal Clinical Sciences, Marion duPont Scott Equine Medical Center , Virginia Tech , Leesburg , VA , USA
| |
Collapse
|
36
|
Kang T, Jones TM, Naddell C, Bacanamwo M, Calvert JW, Thompson WE, Bond VC, Chen YE, Liu D. Adipose-Derived Stem Cells Induce Angiogenesis via Microvesicle Transport of miRNA-31. Stem Cells Transl Med 2016; 5:440-50. [PMID: 26933040 PMCID: PMC4798737 DOI: 10.5966/sctm.2015-0177] [Citation(s) in RCA: 187] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/19/2015] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Cell secretion is an important mechanism for stem cell-based therapeutic angiogenesis, along with cell differentiation to vascular endothelial cells or smooth muscle cells. Cell-released microvesicles (MVs) have been recently implicated to play an essential role in intercellular communication. The purpose of this study was to explore the potential effects of stem cell-released MVs in proangiogenic therapy. We observed for the first time that MVs were released from adipose-derived stem cells (ASCs) and were able to increase the migration and tube formation of human umbilical vein endothelial cells (HUVECs). Endothelial differentiation medium (EDM) preconditioning of ASCs upregulated the release of MVs and enhanced the angiogenic effect of the released MVs in vitro. RNA analysis revealed that microRNA was enriched in ASC-released MVs and that the level of microRNA-31 (miR-31) in MVs was notably elevated upon EDM-preconditioning of MV-donor ASCs. Further studies exhibited that miR-31 in MVs contributed to the migration and tube formation of HUVECs, microvessel outgrowth of mouse aortic rings, and vascular formation of mouse Matrigel plugs. Moreover, factor-inhibiting HIF-1, an antiangiogenic gene, was identified as the target of miR-31 in HUVECs. Our findings provide the first evidence that MVs from ASCs, particularly from EDM-preconditioned ASCs, promote angiogenesis and the delivery of miR-31 may contribute the proangiogenic effect. SIGNIFICANCE This study provides the evidence that microvesicles (MVs) from adipose-derived stem cells (ASCs), particularly from endothelial differentiation medium (EDM)-preconditioned ASCs, promote angiogenesis. An underlying mechanism of the proangiogenesis may be the delivery of microRNA-31 via MVs from ASCs to vascular endothelial cells in which factor-inhibiting HIF-1 is targeted and suppressed. The study findings reveal the role of MVs in mediating ASC-induced angiogenesis and suggest a potential MV-based angiogenic therapy for ischemic diseases.
Collapse
Affiliation(s)
- Ting Kang
- Division of Cardiology, The First Affiliated Hospital, Nanchang University, Nanchang, People's Republic of China Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Tia M Jones
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Clayton Naddell
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Methode Bacanamwo
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - John W Calvert
- Division of Cardiothoracic Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Winston E Thompson
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Vincent C Bond
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Dong Liu
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, USA Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
37
|
Helsley RN, Sui Y, Park SH, Liu Z, Lee RG, Zhu B, Kern PA, Zhou C. Targeting IκB kinase β in Adipocyte Lineage Cells for Treatment of Obesity and Metabolic Dysfunctions. Stem Cells 2016; 34:1883-95. [PMID: 26991836 DOI: 10.1002/stem.2358] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/15/2016] [Indexed: 02/06/2023]
Abstract
IκB kinase β (IKKβ), a central coordinator of inflammation through activation of nuclear factor-κB, has been identified as a potential therapeutic target for the treatment of obesity-associated metabolic dysfunctions. In this study, we evaluated an antisense oligonucleotide (ASO) inhibitor of IKKβ and found that IKKβ ASO ameliorated diet-induced metabolic dysfunctions in mice. Interestingly, IKKβ ASO also inhibited adipocyte differentiation and reduced adiposity in high-fat (HF)-fed mice, indicating an important role of IKKβ signaling in the regulation of adipocyte differentiation. Indeed, CRISPR/Cas9-mediated genomic deletion of IKKβ in 3T3-L1 preadipocytes blocked these cells differentiating into adipocytes. To further elucidate the role of adipose progenitor IKKβ signaling in diet-induced obesity, we generated mice that selectively lack IKKβ in the white adipose lineage and confirmed the essential role of IKKβ in mediating adipocyte differentiation in vivo. Deficiency of IKKβ decreased HF-elicited adipogenesis in addition to reducing inflammation and protected mice from diet-induced obesity and insulin resistance. Further, pharmacological inhibition of IKKβ also blocked human adipose stem cell differentiation. Our findings establish IKKβ as a pivotal regulator of adipogenesis and suggest that overnutrition-mediated IKKβ activation serves as an initial signal that triggers adipose progenitor cell differentiation in response to HF feeding. Inhibition of IKKβ with antisense therapy may represent as a novel therapeutic approach to combat obesity and metabolic dysfunctions. Stem Cells 2016;34:1883-1895.
Collapse
Affiliation(s)
- Robert N Helsley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Yipeng Sui
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Se-Hyung Park
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Zun Liu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Richard G Lee
- Cardiovascular Antisense Drug Discovery Group, Ionis Pharmaceuticals, Inc., Carlsbad, California, USA
| | - Beibei Zhu
- Department of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Philip A Kern
- Department of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Changcheng Zhou
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA.,Saha Cardiovascular Research Center, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
38
|
Siegel-Axel DI, Häring HU. Perivascular adipose tissue: An unique fat compartment relevant for the cardiometabolic syndrome. Rev Endocr Metab Disord 2016; 17:51-60. [PMID: 26995737 DOI: 10.1007/s11154-016-9346-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Type 2 diabetes and its major risk factor, obesity, are an increasing worldwide health problem. The exact mechanisms that link obesity with insulin resistance, type 2 diabetes, hypertension, cardiovascular complications and renal diseases, are still not clarified sufficiently. Adipose tissue in general is an active endocrine and paracrine organ that may influence the development of these disorders. Excessive body fat in general obesity may also cause quantitative and functional alterations of specific adipose tissue compartments. Beside visceral and subcutaneous fat depots which exert systemic effects by the release of adipokines, cytokines and hormones, there are also locally acting fat depots such as peri- and epicardial fat, perivascular fat, and renal sinus fat. Perivascular adipose tissue is in close contact with the adventitia of large, medium and small diameter arteries, possesses unique features differing from other fat depots and may act also independently of general obesity. An increasing number of studies are dealing with the "good" or "bad" characteristics and functions of normally sized and dramatically increased perivascular fat mass in lean or heavily obese individuals. This review describes the origin of perivascular adipose tissue, its different locations, the dual role of a physiological and unphysiological fat mass and its impact on diabetes, cardiovascular and renal diseases. Clinical studies, new imaging methods, as well as basic research in cell culture experiments in the last decade helped to elucidate the various aspects of the unique fat compartment.
Collapse
Affiliation(s)
- D I Siegel-Axel
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University of Tübingen, Tübingen, Germany.
- Institute of Diabetes Research and Metabolic Diseases (IDM), University of Tübingen, Tübingen, Germany.
- Deutsches Zentrum für Diabetesforschung (DZD), Neuherberg, Germany.
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology, and Clinical Chemistry, Eberhard Karls University Tübingen, Otfried-Müller Str.10, D-72076, Tübingen, Germany.
| | - H U Häring
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, University of Tübingen, Tübingen, Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM), University of Tübingen, Tübingen, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), Neuherberg, Germany
| |
Collapse
|
39
|
Wu L, Prins HJ, Leijten J, Helder MN, Evseenko D, Moroni L, van Blitterswijk CA, Lin Y, Karperien M. Chondrocytes Cocultured with Stromal Vascular Fraction of Adipose Tissue Present More Intense Chondrogenic Characteristics Than with Adipose Stem Cells. Tissue Eng Part A 2016; 22:336-48. [PMID: 26732248 DOI: 10.1089/ten.tea.2015.0269] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Partial replacement of chondrocytes by stem cells has been proposed to improve the performance of autologous chondrocyte implantation (ACI). Our previous studies showed that the increased cartilage production in pellet cocultures of chondrocytes and mesenchymal stem cells (MSCs) is due to a trophic role of the MSCs by stimulating chondrocyte proliferation and matrix production rather than MSCs actively undergoing chondrogenic differentiation. The aim of this study is to compare the trophic effects of stromal vascular fraction cells (SVF) and in vitro expanded adipose stem cells (ASC). SVF and culture-expanded ASC (n = 9) were cocultured with primary human chondrocytes in pellets. By glycosaminoglycan (GAG) and DNA assays, we showed that coculture pellets of SVF and chondrocytes have more GAG deposition than that of ASC and chondrocytes. Results of the short tandem repeats analysis indicated that the increase in the chondrocyte proportion in the coculture pellets is more pronounced in the SVF coculture group than in the ASC coculture group. Using flow cytometry and microarray, we demonstrated that SVF and ASC have different characteristics in cell surface markers and gene expression profiles. SVF is more heterogeneous than ASC, whereas ASC is more enriched in cells from the mesenchymal lineage than SVF. By subcutaneous implantation into nude mice, we showed that constructs of SVF and chondrocytes are better in depositing cartilage matrix than the mixture of ASC and chondrocytes. Taken together, SVF is better than ASC in terms of forming cartilage matrix in pellet coculture and in coimplantation models omitting the need for prior cell expansion. Our study suggests that the SVF in combination with primary human chondrocytes may be a good cell combination for one-stage cartilage repair.
Collapse
Affiliation(s)
- Ling Wu
- 1 Department of Developmental BioEngineering, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands .,2 Department of Orthopedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Henk-Jan Prins
- 3 Department of Oral Cell Biology, Academic Center for Dentistry , Amsterdam, The Netherlands .,4 Department of Oral & Maxillofacial Surgery, VU Medical Center , Amsterdam, The Netherlands
| | - Jeroen Leijten
- 1 Department of Developmental BioEngineering, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Marco N Helder
- 5 Department of Orthopedics, VU Medical Center , Amsterdam, The Netherlands
| | - Denis Evseenko
- 2 Department of Orthopedic Surgery, Orthopedic Hospital Research Center, David Geffen School of Medicine, University of California , Los Angeles, California
| | - Lorenzo Moroni
- 6 Department of Tissue Regeneration, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Clemens A van Blitterswijk
- 6 Department of Tissue Regeneration, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Yunfeng Lin
- 7 State Key Laboratory for Oral Diseases, West China School of Stomatology, Sichuan University , Chengdu, China
| | - Marcel Karperien
- 1 Department of Developmental BioEngineering, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| |
Collapse
|
40
|
Song YH, Shon SH, Shan M, Stroock A, Fischbach C. Adipose-derived stem cells increase angiogenesis through matrix metalloproteinase-dependent collagen remodeling. Integr Biol (Camb) 2016; 8:205-15. [PMID: 26758423 PMCID: PMC4755818 DOI: 10.1039/c5ib00277j] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adipose-derived stem cells (ASCs) are key regulators of new blood vessel formation and widely investigated for their role in tissue regeneration and tumorigenesis. However, the cellular and molecular mechanisms through which ASCs regulate angiogenesis are not well understood. Here, it was our goal to test the functional contribution of ASC-mediated extracellular matrix (ECM) remodeling on endothelial cell invasion. To isolate the effect of ECM-remodeling, ASCs were embedded within 3-D collagen type I hydrogels and pre-cultured for 7 days; controls were not pre-cultured. A confluent monolayer of human umbilical vein endothelial cells (HUVECs) was seeded on top and its invasion into the underlying hydrogel was analyzed. Without pre-culture, ASCs inhibited vascular sprouting by stabilizing the endothelium. In contrast, 7 day pre-culture of ASCs drastically increased invasion by HUVECs. This effect was largely mediated by proteolytic ECM degradation by ASC-derived matrix metalloproteinases (MMPs) rather than vascular endothelial growth factor (VEGF), as our results indicated that blockade of MMPs, but not VEGF, inhibited endothelial sprouting. Collectively, these data suggest that the angiogenic capability of ASCs is modulated by their proteolytic remodeling of the ECM, opening new avenues for pro- and anti-angiogenic therapies.
Collapse
Affiliation(s)
- Young Hye Song
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Seung Hee Shon
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Mengrou Shan
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY
| | - Abraham Stroock
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY
| |
Collapse
|
41
|
Klar AS, Güven S, Zimoch J, Zapiórkowska NA, Biedermann T, Böttcher-Haberzeth S, Meuli-Simmen C, Martin I, Scherberich A, Reichmann E, Meuli M. Characterization of vasculogenic potential of human adipose-derived endothelial cells in a three-dimensional vascularized skin substitute. Pediatr Surg Int 2016; 32:17-27. [PMID: 26621500 DOI: 10.1007/s00383-015-3808-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/09/2015] [Indexed: 12/24/2022]
Abstract
PURPOSE The need for clinically applicable skin substitutes continues to be a matter of fact. Hypothetically, a laboratory grown autologous skin analog with near normal architecture might be a suitable approach to yield both satisfactory functional and cosmetic long-term results. In this study, we explored the use of human endothelial cells derived from freshly isolated adipose stromal vascular fraction (SVF) in a three-dimensional (3D) co-culture model of vascularized bio-engineered skin substitute. METHODS The SVF was isolated from human white adipose tissue samples and keratinocytes from human skin biopsies. The SVF, in particular endothelial cells, were characterized using flow cytometry and immuofluorescence analysis. Endothelial and mesenchymal progenitors from the SVF formed blood capillaries after seeding into a 3D collagen type I hydrogel in vitro. Subsequently, human keratinocytes were seeded on the top of those hydrogels to develop a vascularized dermo-epidermal skin substitute. RESULTS Flow cytometric analysis of surface markers of the freshly isolated SVF showed the expression of endothelial markers (CD31, CD34, CD146), mesenchymal/stromal cell-associated markers (CD44, CD73, CD90, CD105), stem cell markers (CD49f, CD117, CD133), and additionally hematopoietic markers (CD14, CD15, CD45). Further analysis of white adipose-derived endothelial cells (watECs) revealed the co-expression of CD31, CD34, CD90, CD105, and partially CD146 on these cells. WatECs were separated from adipose-stromal cells (watASCs) using FACS sorting. WatASCs and watECs cultured separately in a 3D hydrogel for 3 weeks did not form any vascular structures. Only if co-cultured, both cell types aligned to develop a ramified vascular network in vitro with continuous endothelial lumen formation. Transplantation of those 3D-hydrogels onto immuno-incompetent rats resulted in a rapid connection of human capillaries with the host vessels and formation of functional, blood-perfused mosaic human-rat vessels within only 3-4 days. CONCLUSIONS Adipose tissue represents an attractive cell source due to the ease of isolation and abundance of endothelial as well as mesenchymal cell lineages. Adipose-derived SVF cells exhibit the ability to form microvascular structures in vitro and support the accelerated blood perfusion in skin substitutes in vivo when transplanted.
Collapse
Affiliation(s)
- Agnes S Klar
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Sinan Güven
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Jakub Zimoch
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Natalia A Zapiórkowska
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Sophie Böttcher-Haberzeth
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- Department of Surgery, University Children's Hospital Zurich, Steinwiesstrasse 75, 8032, Zurich, Switzerland
| | - Claudia Meuli-Simmen
- Department of Plastic, Reconstructive, Esthetical and Hand Surgery, Kantonsspital Aarau, Aarau, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Department of Biomedicine, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Ernst Reichmann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Martin Meuli
- Department of Surgery, University Children's Hospital Zurich, Steinwiesstrasse 75, 8032, Zurich, Switzerland.
| |
Collapse
|
42
|
Sheng G. The developmental basis of mesenchymal stem/stromal cells (MSCs). BMC DEVELOPMENTAL BIOLOGY 2015; 15:44. [PMID: 26589542 PMCID: PMC4654913 DOI: 10.1186/s12861-015-0094-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 11/13/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Mesenchymal Stem/Stromal Cells (MSCs) define a population of progenitor cells capable of giving rises to at least three mesodermal lineages in vitro, the chondrocytes, osteoblasts and adipocytes. The validity of MSCs in vivo has been questioned because their existence, either as a homogeneous progenitor cell population or as a stem cell lineage, has been difficult to prove. The wide use of primary MSCs in regenerative and therapeutic applications raises ethical and regulatory concerns in many countries. In contrast to hematopoietic stem cells, a parallel concept which carries an embryological emphasis from its outset, MSCs have attracted little interest among developmental biologists and the embryological basis for their existence, or lack thereof, has not been carefully evaluated. METHODS This article provides a brief, embryological overview of these three mesoderm cell lineages and offers a framework of ontological rationales for the potential existence of MSCs in vivo. RESULTS Emphasis is given to the common somatic lateral plate mesoderm origin of the majority of body's adipose and skeletal tissues and of the major sources used for MSC derivation clinically. Support for the MSC hypothesis also comes from a large body of molecular and lineage analysis data in vivo. CONCLUSIONS It is concluded that despite the lack of a definitive proof, the MSC concept has a firm embryological basis and that advances in MSC research can be facilitated by achieving a better integration with developmental biology.
Collapse
Affiliation(s)
- Guojun Sheng
- Sheng Laboratory, International Research Center for Medical Sciences, Kumamoto University, Kumamoto, 860-0811, Japan.
| |
Collapse
|
43
|
Zwierzina ME, Ejaz A, Bitsche M, Blumer MJF, Mitterberger MC, Mattesich M, Amann A, Kaiser A, Pechriggl EJ, Hörl S, Rostek U, Pierer G, Fritsch H, Zwerschke W. Characterization of DLK1(PREF1)+/CD34+ cells in vascular stroma of human white adipose tissue. Stem Cell Res 2015; 15:403-18. [PMID: 26342195 DOI: 10.1016/j.scr.2015.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 08/07/2015] [Accepted: 08/13/2015] [Indexed: 02/07/2023] Open
Abstract
Sorting of native (unpermeabilized) SVF-cells from human subcutaneous (s)WAT for cell surface staining (cs) of DLK1 and CD34 identified three main populations: ~10% stained cs-DLK1+/cs-CD34-, ~20% cs-DLK1+/cs-CD34+dim and ~45% cs-DLK1-/cs-CD34+. FACS analysis after permeabilization showed that all these cells stained positive for intracellular DLK1, while CD34 was undetectable in cs-DLK1+/cs-CD34- cells. Permeabilized cs-DLK1-/cs-CD34+ cells were positive for the pericyte marker α-SMA and the mesenchymal markers CD90 and CD105, albeit CD105 staining was dim (cs-DLK1-/cs-CD34+/CD90+/CD105+dim/α-SMA+/CD45-/CD31-). Only these cells showed proliferative and adipogenic capacity. Cs-DLK1+/cs-CD34- and cs-DLK1+/cs-CD34+dim cells were also α-SMA+ but expressed CD31, had a mixed hematopoietic and mesenchymal phenotype, and could neither proliferate nor differentiate into adipocytes. Histological analysis of sWAT detected DLK1+/CD34+ and DLK1+/CD90+ cells mainly in the outer ring of vessel-associated stroma and at capillaries. DLK1+/α-SMA+ cells were localized in the CD34- perivascular ring and in adventitial vascular stroma. All these DLK1+ cells possess a spindle-shaped morphology with extremely long processes. DLK1+/CD34+ cells were also detected in vessel endothelium. Additionally, we show that sWAT contains significantly more DLK1+ cells than visceral (v)WAT. We conclude that sWAT has more DKL1+ cells than vWAT and contains different DLK1/CD34 populations, and only cs-DLK1-/cs-CD34+/CD90+/CD105+dim/α-SMA+/CD45-/CD31- cells in the adventitial vascular stroma exhibit proliferative and adipogenic capacity.
Collapse
Affiliation(s)
- Marit E Zwierzina
- Division for Clinical and Functional Anatomy, Department for Anatomy, Histology and Embryology, Medical University of Innsbruck, Austria
| | - Asim Ejaz
- Cell Metabolism and Differentiation Research Group, Institute for Biomedical Aging Research, University of Innsbruck, Austria
| | - Mario Bitsche
- Division for Clinical and Functional Anatomy, Department for Anatomy, Histology and Embryology, Medical University of Innsbruck, Austria
| | - Michael J F Blumer
- Division for Clinical and Functional Anatomy, Department for Anatomy, Histology and Embryology, Medical University of Innsbruck, Austria
| | - Maria C Mitterberger
- Cell Metabolism and Differentiation Research Group, Institute for Biomedical Aging Research, University of Innsbruck, Austria
| | - Monika Mattesich
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Austria
| | - Arno Amann
- Department of Internal Medicine V, Medical University of Innsbruck, Austria
| | - Andreas Kaiser
- Cell Metabolism and Differentiation Research Group, Institute for Biomedical Aging Research, University of Innsbruck, Austria
| | - Elisabeth J Pechriggl
- Division for Clinical and Functional Anatomy, Department for Anatomy, Histology and Embryology, Medical University of Innsbruck, Austria
| | - Susanne Hörl
- Cell Metabolism and Differentiation Research Group, Institute for Biomedical Aging Research, University of Innsbruck, Austria
| | - Ursula Rostek
- Cell Metabolism and Differentiation Research Group, Institute for Biomedical Aging Research, University of Innsbruck, Austria
| | - Gerhard Pierer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Austria
| | - Helga Fritsch
- Division for Clinical and Functional Anatomy, Department for Anatomy, Histology and Embryology, Medical University of Innsbruck, Austria
| | - Werner Zwerschke
- Cell Metabolism and Differentiation Research Group, Institute for Biomedical Aging Research, University of Innsbruck, Austria.
| |
Collapse
|
44
|
Abstract
Regenerative capacity of skeletal muscles resides in satellite cells, a self-renewing population of muscle cells. Several studies are investigating epigenetic mechanisms that control myogenic proliferation and differentiation to find new approaches that could boost regeneration of endogenous myogenic progenitor populations. In recent years, a lot of effort has been applied to purify, expand and manipulate adult stem cells from muscle tissue. However, this population of endogenous myogenic progenitors in adults is limited and their access is difficult and invasive. Therefore, other sources of stem cells with potential to regenerate muscles need to be examined. An excellent candidate could be a population of adult stromal cells within fat characterized by mesenchymal properties, which have been termed adipose-derived stem cells (ASCs). These progenitor adult stem cells have been successfully differentiated in vitro to osteogenic, chondrogenic, neurogenic and myogenic lineages. Autologous ASCs are multipotent and can be harvested with low morbidity; thus, they hold promise for a range of therapeutic applications. This review will summarize the use of ASCs in muscle regenerative approaches.
Collapse
Affiliation(s)
- Sonia-V Forcales
- Genetics and Epigenetics of Cancer, Institute of Predictive and Personalized Medicine of Cancer Barcelona, Spain
| |
Collapse
|
45
|
Cronk SM, Kelly-Goss MR, Ray HC, Mendel TA, Hoehn KL, Bruce AC, Dey BK, Guendel AM, Tavakol DN, Herman IM, Peirce SM, Yates PA. Adipose-derived stem cells from diabetic mice show impaired vascular stabilization in a murine model of diabetic retinopathy. Stem Cells Transl Med 2015; 4:459-67. [PMID: 25769654 DOI: 10.5966/sctm.2014-0108] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 02/09/2015] [Indexed: 12/15/2022] Open
Abstract
Diabetic retinopathy is characterized by progressive vascular dropout with subsequent vision loss. We have recently shown that an intravitreal injection of adipose-derived stem cells (ASCs) can stabilize the retinal microvasculature, enabling repair and regeneration of damaged capillary beds in vivo. Because an understanding of ASC status from healthy versus diseased donors will be important as autologous cellular therapies are developed for unmet clinical needs, we took advantage of the hyperglycemic Akimba mouse as a preclinical in vivo model of diabetic retinopathy in an effort aimed at evaluating therapeutic efficacy of adipose-derived stem cells (mASCs) derived either from healthy, nondiabetic or from diabetic mice. To these ends, Akimba mice received intravitreal injections of media conditioned by mASCs or mASCs themselves, subsequent to development of substantial retinal capillary dropout. mASCs from healthy mice were more effective than diabetic mASCs in protecting the diabetic retina from further vascular dropout. Engrafted ASCs were found to preferentially associate with the retinal vasculature. Conditioned medium was unable to recapitulate the vasoprotection seen with injected ASCs. In vitro diabetic ASCs showed decreased proliferation and increased apoptosis compared with healthy mASCs. Diabetic ASCs also secreted less vasoprotective factors than healthy mASCs, as determined by high-throughput enzyme-linked immunosorbent assay. Our findings suggest that diabetic ASCs are functionally impaired compared with healthy ASCs and support the utility of an allogeneic injection of ASCs versus autologous or conditioned media approaches in the treatment of diabetic retinopathy.
Collapse
Affiliation(s)
- Stephen M Cronk
- Departments of Biomedical Engineering, Pathology, Pharmacology, and Ophthalmology, University of Virginia, Charlottesville, Virginia, USA; Department of Developmental, Molecular and Chemical Biology and Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, Boston, Massachusetts, USA
| | - Molly R Kelly-Goss
- Departments of Biomedical Engineering, Pathology, Pharmacology, and Ophthalmology, University of Virginia, Charlottesville, Virginia, USA; Department of Developmental, Molecular and Chemical Biology and Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, Boston, Massachusetts, USA
| | - H Clifton Ray
- Departments of Biomedical Engineering, Pathology, Pharmacology, and Ophthalmology, University of Virginia, Charlottesville, Virginia, USA; Department of Developmental, Molecular and Chemical Biology and Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, Boston, Massachusetts, USA
| | - Thomas A Mendel
- Departments of Biomedical Engineering, Pathology, Pharmacology, and Ophthalmology, University of Virginia, Charlottesville, Virginia, USA; Department of Developmental, Molecular and Chemical Biology and Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, Boston, Massachusetts, USA
| | - Kyle L Hoehn
- Departments of Biomedical Engineering, Pathology, Pharmacology, and Ophthalmology, University of Virginia, Charlottesville, Virginia, USA; Department of Developmental, Molecular and Chemical Biology and Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, Boston, Massachusetts, USA
| | - Anthony C Bruce
- Departments of Biomedical Engineering, Pathology, Pharmacology, and Ophthalmology, University of Virginia, Charlottesville, Virginia, USA; Department of Developmental, Molecular and Chemical Biology and Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, Boston, Massachusetts, USA
| | - Bijan K Dey
- Departments of Biomedical Engineering, Pathology, Pharmacology, and Ophthalmology, University of Virginia, Charlottesville, Virginia, USA; Department of Developmental, Molecular and Chemical Biology and Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, Boston, Massachusetts, USA
| | - Alexander M Guendel
- Departments of Biomedical Engineering, Pathology, Pharmacology, and Ophthalmology, University of Virginia, Charlottesville, Virginia, USA; Department of Developmental, Molecular and Chemical Biology and Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, Boston, Massachusetts, USA
| | - Daniel N Tavakol
- Departments of Biomedical Engineering, Pathology, Pharmacology, and Ophthalmology, University of Virginia, Charlottesville, Virginia, USA; Department of Developmental, Molecular and Chemical Biology and Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, Boston, Massachusetts, USA
| | - Ira M Herman
- Departments of Biomedical Engineering, Pathology, Pharmacology, and Ophthalmology, University of Virginia, Charlottesville, Virginia, USA; Department of Developmental, Molecular and Chemical Biology and Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, Boston, Massachusetts, USA
| | - Shayn M Peirce
- Departments of Biomedical Engineering, Pathology, Pharmacology, and Ophthalmology, University of Virginia, Charlottesville, Virginia, USA; Department of Developmental, Molecular and Chemical Biology and Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, Boston, Massachusetts, USA
| | - Paul A Yates
- Departments of Biomedical Engineering, Pathology, Pharmacology, and Ophthalmology, University of Virginia, Charlottesville, Virginia, USA; Department of Developmental, Molecular and Chemical Biology and Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
46
|
Mortensen LJ, Alt C, Turcotte R, Masek M, Liu TM, Côté DC, Xu C, Intini G, Lin CP. Femtosecond laser bone ablation with a high repetition rate fiber laser source. BIOMEDICAL OPTICS EXPRESS 2015; 6:32-42. [PMID: 25657872 PMCID: PMC4317121 DOI: 10.1364/boe.6.000032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 09/29/2014] [Accepted: 09/30/2014] [Indexed: 05/20/2023]
Abstract
Femtosecond laser pulses can be used to perform very precise cutting of material, including biological samples from subcellular organelles to large areas of bone, through plasma-mediated ablation. The use of a kilohertz regenerative amplifier is usually needed to obtain the pulse energy required for ablation. This work investigates a 5 megahertz compact fiber laser for near-video rate imaging and ablation in bone. After optimization of ablation efficiency and reduction in autofluorescence, the system is demonstrated for the in vivo study of bone regeneration. Image-guided creation of a bone defect and longitudinal evaluation of cellular injury response in the defect provides insight into the bone regeneration process.
Collapse
Affiliation(s)
- Luke J. Mortensen
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts,
USA
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts,
USA
| | - Clemens Alt
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts,
USA
| | - Raphaël Turcotte
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts,
USA
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts,
USA
| | - Marissa Masek
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts,
USA
| | - Tzu-Ming Liu
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts,
USA
- Institute of Biomedical Engineering, National Taiwan University, Taipei,
Taiwan
| | - Daniel C. Côté
- Centre de Recherche Université Laval Robert-Giffard, Université Laval, Québec, QC G1J2G3,
Canada
| | - Chris Xu
- School of Applied and Engineering Physics, Cornell University, Ithaca, New York,
USA
| | - Giuseppe Intini
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts,
USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts,
USA
- Co-corresponding authors
| | - Charles P. Lin
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts,
USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts,
USA
- Co-corresponding authors
| |
Collapse
|
47
|
Birbrair A, Zhang T, Wang ZM, Messi ML, Mintz A, Delbono O. Pericytes at the intersection between tissue regeneration and pathology. Clin Sci (Lond) 2015; 128:81-93. [PMID: 25236972 PMCID: PMC4200531 DOI: 10.1042/cs20140278] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Perivascular multipotent cells, pericytes, contribute to the generation and repair of various tissues in response to injury. They are heterogeneous in their morphology, distribution, origin and markers, and elucidating their molecular and cellular differences may inform novel treatments for disorders in which tissue regeneration is either impaired or excessive. Moreover, these discoveries offer novel cellular targets for therapeutic approaches to many diseases. This review discusses recent studies that support the concept that pericyte subtypes play a distinctive role in myogenesis, neurogenesis, adipogenesis, fibrogenesis and angiogenesis.
Collapse
Affiliation(s)
- Alexander Birbrair
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Tan Zhang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Zhong-Min Wang
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Maria Laura Messi
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Akiva Mintz
- Department of Neurosurgery, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| | - Osvaldo Delbono
- Department of Internal Medicine-Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina, Medical Center Boulevard, Winston Salem, NC 27157, U.S.A
| |
Collapse
|
48
|
Haug V, Torio-Padron N, Stark GB, Finkenzeller G, Strassburg S. Comparison between endothelial progenitor cells and human umbilical vein endothelial cells on neovascularization in an adipogenesis mouse model. Microvasc Res 2015; 97:159-66. [DOI: 10.1016/j.mvr.2014.10.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/15/2014] [Accepted: 10/17/2014] [Indexed: 01/22/2023]
|
49
|
Abstract
Context: Articular cartilage possesses poor natural healing mechanisms, and a variety of non-cell-based and cell-based treatments aim to promote regeneration of hyaline cartilage. Data Sources: A review of the literature to December 2013 using PubMed with search criteria including the keywords stem cell, cell therapy, cell transplantation, cartilage, chondral, and chondrogenic. Study Selection: Forty-five articles were identified that employed local mesenchymal stem cell (MSC) therapy for joint disorders in humans. Nine comparative studies were identified, consisting of 3 randomized trials, 5 cohort studies, and 1 case-control study. Study Type: Clinical review. Level of Evidence: Level 4. Data Extraction: Studies were assessed for stem cell source, method of implantation, comparison groups, and concurrent surgical techniques. Results: Two studies comparing MSC treatment to autologous chondrocyte implantation found similar efficacy. Three studies reported clinical benefits with intra-articular MSC injection over non-MSC controls for cases undergoing debridement with or without marrow stimulation, although a randomized study found no significant clinical difference at 2-year follow-up but reported better 18-month magnetic resonance imaging and histologic scores in the MSC group. No human studies have compared intra-articular MSC therapy to non-MSC techniques for osteoarthritis in the absence of surgery. Conclusion: Mesenchymal stem cell–based therapies appear safe and effective for joint disorders in large animal preclinical models. Evidence for use in humans, particularly, comparison with more established treatments such as autologous chondrocyte implantation and microfracture, is limited.
Collapse
Affiliation(s)
| | - Daniel Bates
- Lakeside Sports Medicine Centre, Melbourne, Victoria, Australia
| | - Richard Boyd
- Monash Immunology and Stem Cell Laboratories (MISCL), Monash University, Clayton, Victoria, Australia
| | - David A Connell
- Faculty of Medicine, Nursing & Healthcare, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
50
|
Comparative analysis of cardiovascular development related genes in stem cells isolated from deciduous pulp and adipose tissue. ScientificWorldJournal 2014; 2014:186508. [PMID: 25548778 PMCID: PMC4273554 DOI: 10.1155/2014/186508] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 10/20/2014] [Accepted: 11/03/2014] [Indexed: 12/30/2022] Open
Abstract
Human exfoliated deciduous teeth (SHED) and adipose stem cells (ASC) were suggested as alternative cell choice for cardiac regeneration. However, the true functionability of these cells toward cardiac regeneration is yet to be discovered. Hence, this study was carried out to investigate the innate biological properties of these cell sources toward cardiac regeneration. Both cells exhibited indistinguishable MSCs characteristics. Human stem cell transcription factor arrays were used to screen expression levels in SHED and ASC. Upregulated expression of transcription factor (TF) genes was detected in both sources. An almost equal percentage of > 2-fold changes were observed. These TF genes fall under several cardiovascular categories with higher expressions which were observed in growth and development of blood vessel, angiogenesis, and vasculogenesis categories. Further induction into cardiomyocyte revealed ASC to express more significantly cardiomyocyte specific markers compared to SHED during the differentiation course evidenced by morphology and gene expression profile. Despite this, spontaneous cellular beating was not detected in both cell lines. Taken together, our data suggest that despite being defined as MSCs, both ASC and SHED behave differently when they were cultured in a same cardiomyocytes culture condition. Hence, vigorous characterization is needed before introducing any cell for treating targeted diseases.
Collapse
|