1
|
Pakdaman Kolour SS, Nematollahi S, Dehbozorgi M, Fattahi F, Movahed F, Esfandiari N, Kahrizi MS, Ghavamikia N, Hajiagha BS. Extracecellulr vesicles (EVs) microRNAs (miRNAs) derived from mesenchymal stem cells (MSCs) in osteoarthritis (OA); detailed role in pathogenesis and possible therapeutics. Heliyon 2025; 11:e42258. [PMID: 40007782 PMCID: PMC11850152 DOI: 10.1016/j.heliyon.2025.e42258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
The primary cause of pain and disability in the world is osteoarthritis (OA), a common joint disease characterized by the primary pathological alteration in articular cartilage deterioration. The general outcome of treatment is not acceptable despite current interventions. Therefore, joint replacement surgery is frequently needed by patients with severe OA. Mesenchymal stem cells (MSCs) have become a practical treatment choice for preclinical and clinical OA palliation in recent years, mainly due to their unique immunomodulatory attributes. Further, attractive candidates for cell-free therapy for OA are MSC-derived extracecellulr vesicles (EVs) that convey bioactive molecules of the original cells, such as microRNAs. These EVs have been shown to significantly influence the regulation of various physiological activities of cells in the joint cavity. Dysregulated miRNAs upregulate the synthesis of enzymes that degrade cartilage, downregulate the expression of components in the cartilage matrix, promote the production of proinflammatory cytokines, induce programmed cell death in chondrocytes, inhibit the process of autophagy in chondrocytes, and participate in pathways related to pain. MiRNAs are also found in extracellular membranous vesicles (EVs), such as exosomes, and play a role in intercellular communication in osteoarthritic joints. Thus, the biosynthesis, chemical makeup, and mechanism of action of miRNAs-enriched EVs in OA are all thoroughly covered in this review. We additionally discussed how miRNA-enriched MSC-EVs might be used therapeutically to change intercellular interaction in OA.
Collapse
Affiliation(s)
| | - Saeide Nematollahi
- Department of Radiology, Kerman University of Paramedical Sciences, Kerman, Iran
| | | | | | - Fatemeh Movahed
- Department of Gynecology, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Nima Ghavamikia
- Cardiovascular Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Salmanian Hajiagha
- Department of Cellular and Molecular Biology, Faculty of Basic Science, East Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
2
|
Lempesis IG, Hoebers N, Essers Y, Jocken JWE, Dubois LJ, Blaak EE, Manolopoulos KN, Goossens GH. Impaired Mitochondrial Respiration in Upper Compared to Lower Body Differentiated Human Adipocytes and Adipose Tissue. J Clin Endocrinol Metab 2024; 109:e2291-e2301. [PMID: 38375937 PMCID: PMC11570378 DOI: 10.1210/clinem/dgae086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 01/30/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
CONTEXT Abdominal obesity is associated with increased cardiometabolic disease risk, while lower body fat seems to confer protection against obesity-related complications. The functional differences between upper and lower body adipose tissue (AT) remain poorly understood. OBJECTIVE We aimed to examine whether mitochondrial respiration is impaired in abdominal as compared to femoral differentiated human multipotent adipose-derived stem cells (hMADS; primary outcome) and AT in postmenopausal women. DESIGN In this cross-sectional study, 23 postmenopausal women with normal weight or obesity were recruited at the University of Birmingham/Queen Elizabeth Hospital Birmingham (Birmingham, UK). We collected abdominal and femoral subcutaneous AT biopsies to determine mitochondrial oxygen consumption rates in differentiated abdominal and femoral hMADS. Furthermore, we assessed oxidative phosphorylation (OXPHOS) protein expression and mitochondrial DNA (mtDNA) content in abdominal and femoral AT as well as hMADS. Finally, we explored in vivo fractional oxygen extraction and carbon dioxide release across abdominal and femoral subcutaneous AT in a subgroup of the same individuals with normal weight or obesity. RESULTS We found lower basal and maximal uncoupled mitochondrial oxygen consumption rates in abdominal compared to femoral hMADS. In line, in vivo fractional oxygen extraction and carbon dioxide release were lower across abdominal than femoral AT. OXPHOS protein expression and mtDNA content did not significantly differ between abdominal and femoral differentiated hMADS and AT. CONCLUSION The present findings demonstrate that in vitro mitochondrial respiration and in vivo oxygen fractional extraction are less in upper compared to lower body differentiated hMADS and AT, respectively, in postmenopausal women.
Collapse
Affiliation(s)
- Ioannis G Lempesis
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200 MD Maastricht, The Netherlands
| | - Nicole Hoebers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200 MD Maastricht, The Netherlands
| | - Yvonne Essers
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200 MD Maastricht, The Netherlands
| | - Johan W E Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200 MD Maastricht, The Netherlands
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW School for Oncology and Reproduction, Maastricht University Medical Centre+, 6200 MD Maastricht, The Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200 MD Maastricht, The Netherlands
| | - Konstantinos N Manolopoulos
- Institute of Metabolism and Systems Research (IMSR), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
3
|
Ding Z, Yan Z, Yuan X, Tian G, Wu J, Fu L, Yin H, He S, Ning C, Zheng Y, Zhang Z, Sui X, Hao L, Niu Y, Liu S, Guo W, Guo Q. Apoptotic extracellular vesicles derived from hypoxia-preconditioned mesenchymal stem cells within a modified gelatine hydrogel promote osteochondral regeneration by enhancing stem cell activity and regulating immunity. J Nanobiotechnology 2024; 22:74. [PMID: 38395929 PMCID: PMC10885680 DOI: 10.1186/s12951-024-02333-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Due to its unique structure, articular cartilage has limited abilities to undergo self-repair after injury. Additionally, the repair of articular cartilage after injury has always been a difficult problem in the field of sports medicine. Previous studies have shown that the therapeutic use of mesenchymal stem cells (MSCs) and their extracellular vesicles (EVs) has great potential for promoting cartilage repair. Recent studies have demonstrated that most transplanted stem cells undergo apoptosis in vivo, and the apoptotic EVs (ApoEVs) that are subsequently generated play crucial roles in tissue repair. Additionally, MSCs are known to exist under low-oxygen conditions in the physiological environment, and these hypoxic conditions can alter the functional and secretory properties of MSCs as well as their secretomes. This study aimed to investigate whether ApoEVs that are isolated from adipose-derived MSCs cultured under hypoxic conditions (hypoxic apoptotic EVs [H-ApoEVs]) exert greater effects on cartilage repair than those that are isolated from cells cultured under normoxic conditions. Through in vitro cell proliferation and migration experiments, we demonstrated that H-ApoEVs exerted enhanced effects on stem cell proliferation, stem cell migration, and bone marrow derived macrophages (BMDMs) M2 polarization compared to ApoEVs. Furthermore, we utilized a modified gelatine matrix/3D-printed extracellular matrix (ECM) scaffold complex as a carrier to deliver H-ApoEVs into the joint cavity, thus establishing a cartilage regeneration system. The 3D-printed ECM scaffold provided mechanical support and created a microenvironment that was conducive to cartilage regeneration, and the H-ApoEVs further enhanced the regenerative capacity of endogenous stem cells and the immunomodulatory microenvironment of the joint cavity; thus, this approach significantly promoted cartilage repair. In conclusion, this study confirmed that a ApoEVs delivery system based on a modified gelatine matrix/3D-printed ECM scaffold together with hypoxic preconditioning enhances the functionality of stem cell-derived ApoEVs and represents a promising approach for promoting cartilage regeneration.
Collapse
Affiliation(s)
- Zhengang Ding
- Guizhou Medical University, Guiyang, 550004, Guizhou, China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Zineng Yan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xun Yuan
- Guizhou Medical University, Guiyang, 550004, Guizhou, China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Guangzhao Tian
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jiang Wu
- Guizhou Medical University, Guiyang, 550004, Guizhou, China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Liwei Fu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Han Yin
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Songlin He
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chao Ning
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yazhe Zheng
- Guizhou Medical University, Guiyang, 550004, Guizhou, China
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Zhichao Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiang Sui
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Libo Hao
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yuting Niu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, People's Republic of China.
| | - Shuyun Liu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Weimin Guo
- Department of Orthopaedic Surgery Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
| | - Quanyi Guo
- Guizhou Medical University, Guiyang, 550004, Guizhou, China.
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
4
|
Alemany M. The Metabolic Syndrome, a Human Disease. Int J Mol Sci 2024; 25:2251. [PMID: 38396928 PMCID: PMC10888680 DOI: 10.3390/ijms25042251] [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: 12/01/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
This review focuses on the question of metabolic syndrome (MS) being a complex, but essentially monophyletic, galaxy of associated diseases/disorders, or just a syndrome of related but rather independent pathologies. The human nature of MS (its exceptionality in Nature and its close interdependence with human action and evolution) is presented and discussed. The text also describes the close interdependence of its components, with special emphasis on the description of their interrelations (including their syndromic development and recruitment), as well as their consequences upon energy handling and partition. The main theories on MS's origin and development are presented in relation to hepatic steatosis, type 2 diabetes, and obesity, but encompass most of the MS components described so far. The differential effects of sex and its biological consequences are considered under the light of human social needs and evolution, which are also directly related to MS epidemiology, severity, and relations with senescence. The triggering and maintenance factors of MS are discussed, with especial emphasis on inflammation, a complex process affecting different levels of organization and which is a critical element for MS development. Inflammation is also related to the operation of connective tissue (including the adipose organ) and the widely studied and acknowledged influence of diet. The role of diet composition, including the transcendence of the anaplerotic maintenance of the Krebs cycle from dietary amino acid supply (and its timing), is developed in the context of testosterone and β-estradiol control of the insulin-glycaemia hepatic core system of carbohydrate-triacylglycerol energy handling. The high probability of MS acting as a unique complex biological control system (essentially monophyletic) is presented, together with additional perspectives/considerations on the treatment of this 'very' human disease.
Collapse
Affiliation(s)
- Marià Alemany
- Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
5
|
Engin A. Adipose Tissue Hypoxia in Obesity: Clinical Reappraisal of Hypoxia Hypothesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:329-356. [PMID: 39287857 DOI: 10.1007/978-3-031-63657-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Obese subjects exhibit lower adipose tissue oxygen consumption in accordance with the lower adipose tissue blood flow. Thereby, compared to lean subjects, obese individuals have almost half lower capillary density and more than half lower vascular endothelial growth factor (VEGF). The VEGF expression together with hypoxia-inducible transcription factor-1 alpha (HIF-1α) activity also requires phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR)-mediated signaling. Especially HIF-1α is an important signaling molecule for hypoxia to induce the inflammatory responses. Hypoxia contributes to several biological functions, such as angiogenesis, cell proliferation, apoptosis, inflammation, and insulin resistance (IR). Pathogenesis of obesity-related comorbidities is attributed to intermittent hypoxia (IH), which is mostly observed in visceral obesity. Proinflammatory phenotype of the adipose tissue is a crucial link between IH and the development of IR. Inhibition of adaptive unfolded protein response (UPR) in hypoxia increases β cell death. Moreover, deletion of HIF-1α worsens β cell function. Oxidative stress, as well as the release of proinflammatory cytokines/adipokines in obesity, is proportional to the severity of IH. Reactive oxygen species (ROS) generation at mitochondria is responsible for propagation of the hypoxic signal; however, mitochondrial ROS production is required for hypoxic HIF-1α protein stabilization. Alterations in oxygen availability of adipose tissue directly affect the macrophage polarization and are responsible for the dysregulated adipocytokines production in obesity. Hypoxia both inhibits adipocyte differentiation from preadipocytes and macrophage migration from the hypoxic adipose tissue. Upon reaching a hypertrophic threshold beyond the adipocyte fat loading capacity, excess extracellular matrix (ECM) components are deposited, causing fibrosis. HIF-1α initiates the whole pathological process of fibrosis and inflammation in the obese adipose tissue. In addition to stressed adipocytes, hypoxia contributes to immune cell migration and activation which further aggravates adipose tissue fibrosis. Therefore, targeting HIF-1α might be an efficient way to suppress hypoxia-induced pathological changes in the ECM. The fibrosis score of adipose tissue correlates negatively with the body mass index and metabolic parameters. Inducers of browning/beiging adipocytes and adipokines, as well as modulations of matrix remodeling enzyme inhibitors, and associated gene regulators, are potential pharmacological targets for treating obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
6
|
Kim DH, Kim SH, Park SH, Kwon MY, Lim CY, Park SH, Gwon K, Hwang SH, Kim SW. Characteristics of Human Nasal Turbinate Stem Cells under Hypoxic Conditions. Cells 2023; 12:2360. [PMID: 37830573 PMCID: PMC10571865 DOI: 10.3390/cells12192360] [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: 08/31/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
This study investigated the influence of hypoxic culture conditions on human nasal inferior turbinate-derived stem cells (hNTSCs), a subtype of mesenchymal stem cells (MSCs). It aimed to discern how hypoxia affected hNTSC characteristics, proliferation, and differentiation potential compared to hNTSCs cultured under normal oxygen levels. After obtaining hNTSCs from five patients, the samples were divided into hypoxic and normoxic groups. The investigation utilized fluorescence-activated cell sorting (FACS) for surface marker analysis, cell counting kit-8 assays for proliferation assessment, and multiplex immunoassays for cytokine secretion study. Differentiation potential-osteogenic, chondrogenic, and adipogenic-was evaluated via histological examination and gene expression analysis. Results indicated that hNTSCs under hypoxic conditions preserved their characteristic MSC phenotype, as confirmed by FACS analysis demonstrating the absence of hematopoietic markers and presence of MSC markers. Proliferation of hNTSCs remained unaffected by hypoxia. Cytokine expression showed similarity between hypoxic and normoxic groups throughout cultivation. Nevertheless, hypoxic conditions reduced the osteogenic and promoted adipogenic differentiation potential, while chondrogenic differentiation was relatively unchanged. These insights contribute to understanding hNTSC behavior in hypoxic environments, advancing the development of protocols for stem cell therapies and tissue engineering.
Collapse
Affiliation(s)
- Do Hyun Kim
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.K.); (S.H.K.)
| | - Sun Hong Kim
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.K.); (S.H.K.)
| | - Sang Hi Park
- Institute of Clinical Medicine Research, College of Medicine, Catholic University of Korea, Seoul 06591, Republic of Korea; (S.H.P.); (M.Y.K.); (C.-Y.L.)
| | - Mi Yeon Kwon
- Institute of Clinical Medicine Research, College of Medicine, Catholic University of Korea, Seoul 06591, Republic of Korea; (S.H.P.); (M.Y.K.); (C.-Y.L.)
| | - Chae-Yoon Lim
- Institute of Clinical Medicine Research, College of Medicine, Catholic University of Korea, Seoul 06591, Republic of Korea; (S.H.P.); (M.Y.K.); (C.-Y.L.)
| | - Sun Hwa Park
- Postech-Catholic Biomedical Engineering Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Kihak Gwon
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55902, USA;
| | - Se Hwan Hwang
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.K.); (S.H.K.)
| | - Sung Won Kim
- Department of Otolaryngology—Head and Neck Surgery, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.K.); (S.H.K.)
| |
Collapse
|
7
|
Major G, Longoni A, Simcock J, Magon NJ, Harte J, Bathish B, Kemp R, Woodfield T, Lim KS. Clinical Applicability of Visible Light-Mediated Cross-linking for Structural Soft Tissue Reconstruction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300538. [PMID: 37424046 PMCID: PMC10502829 DOI: 10.1002/advs.202300538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/28/2023] [Indexed: 07/11/2023]
Abstract
Visible light-mediated cross-linking has utility for enhancing the structural capacity and shape fidelity of laboratory-based polymers. With increased light penetration and cross-linking speed, there is opportunity to extend future applications into clinical spheres. This study evaluated the utility of a ruthenium/sodium persulfate photocross-linking system for increasing structural control in heterogeneous living tissues as an example, focusing on unmodified patient-derived lipoaspirate for soft tissue reconstruction. Freshly-isolated tissue is photocross-linked, then the molar abundance of dityrosine bonds is measured using liquid chromatography tandem mass spectrometry and the resulting structural integrity assessed. The cell function and tissue survival of photocross-linked grafts is evaluated ex vivo and in vivo, with tissue integration and vascularization assessed using histology and microcomputed tomography. The photocross-linking strategy is tailorable, allowing progressive increases in the structural fidelity of lipoaspirate, as measured by a stepwise reduction in fiber diameter, increased graft porosity and reduced variation in graft resorption. There is an increase in dityrosine bond formation with increasing photoinitiator concentration, and tissue homeostasis is achieved ex vivo, with vascular cell infiltration and vessel formation in vivo. These data demonstrate the capability and applicability of photocrosslinking strategies for improving structural control in clinically-relevant settings, potentially achieving more desirable patient outcomes using minimal manipulation in surgical procedures.
Collapse
Affiliation(s)
- Gretel Major
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
| | - Alessia Longoni
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
| | - Jeremy Simcock
- Department of SurgeryUniversity of OtagoChristchurch8011New Zealand
| | - Nicholas J Magon
- Centre for Free Radical ResearchDepartment of Pathology and Biomedical ScienceUniversity of OtagoChristchurch8011New Zealand
| | - Jessica Harte
- Jacqui Wood Cancer CentreDivision of Cellular MedicineNinewells Hospital and Medical SchoolUniversity of DundeeDundeeScotlandDD2 1GZUK
| | - Boushra Bathish
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
- Jacqui Wood Cancer CentreDivision of Cellular MedicineNinewells Hospital and Medical SchoolUniversity of DundeeDundeeScotlandDD2 1GZUK
| | - Roslyn Kemp
- Department of Microbiology and ImmunologyUniversity of OtagoDunedin9016New Zealand
| | - Tim Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
| | - Khoon S Lim
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
- Light‐Activated Biomaterials GroupSchool of Medical SciencesUniversity of SydneySydney2006Australia
| |
Collapse
|
8
|
Yang Y, Wu Y, Yang D, Neo SH, Kadir ND, Goh D, Tan JX, Denslin V, Lee EH, Yang Z. Secretive derived from hypoxia preconditioned mesenchymal stem cells promote cartilage regeneration and mitigate joint inflammation via extracellular vesicles. Bioact Mater 2023; 27:98-112. [PMID: 37006826 PMCID: PMC10063382 DOI: 10.1016/j.bioactmat.2023.03.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/23/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Secretome derived from mesenchymal stem cells (MSCs) have profound effects on tissue regeneration, which could become the basis of future MSCs therapies. Hypoxia, as the physiologic environment of MSCs, has great potential to enhance MSCs paracrine therapeutic effect. In our study, the paracrine effects of secretome derived from MSCs preconditioned in normoxia and hypoxia was compared through both in vitro functional assays and an in vivo rat osteochondral defect model. Specifically, the paracrine effect of total EVs were compared to that of soluble factors to characterize the predominant active components in the hypoxic secretome. We demonstrated that hypoxia conditioned medium, as well as the corresponding EVs, at a relatively low dosage, were efficient in promoting the repair of critical-sized osteochondral defects and mitigated the joint inflammation in a rat osteochondral defect model, relative to their normoxia counterpart. In vitro functional test shows enhancement through chondrocyte proliferation, migration, and matrix deposition, while inhibit IL-1β-induced chondrocytes senescence, inflammation, matrix degradation, and pro-inflammatory macrophage activity. Multiple functional proteins, as well as a change in EVs' size profile, with enrichment of specific EV-miRNAs were detected with hypoxia preconditioning, implicating complex molecular pathways involved in hypoxia pre-conditioned MSCs secretome generated cartilage regeneration.
Collapse
|
9
|
Dong J, Wu B, Tian W. Exosomes derived from hypoxia-preconditioned mesenchymal stem cells (hypoMSCs-Exo): advantages in disease treatment. Cell Tissue Res 2023:10.1007/s00441-023-03758-6. [PMID: 36781483 DOI: 10.1007/s00441-023-03758-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/04/2023] [Indexed: 02/15/2023]
Abstract
Mesenchymal stem cells (MSCs)-based therapy has been reported to be a potential approach to treat various diseases and the paracrine role might be the underlying mechanism. Exosomes were considered an important part of this paracrine role. It was reported that maintenance of MSCs in hypoxia conditions for a short time has shown to be beneficial for the therapeutic effect of MSCs and MSCs-derived exosomes. In this review, we summarized the recent developments on exosomes derived from hypoxia-preconditioned mesenchymal stem cells (hypoMSCs-Exo), including the characteristics of hypoMSCs-Exo in morphology and contents, diseases in which hypoMSCs-Exo showed more effective, and the cellular and molecular mechanisms that hypoMSCs-Exo showed more effective in disease treatment. Besides, we also discussed the limitations of current studies and the issues that needed to be improved in the application of hypoMSCs-Exo. This review aimed to promote a comprehensive and systematic understanding of this type of exosome with great therapeutic potential.
Collapse
Affiliation(s)
- Jia Dong
- State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China. .,Department of Stomatology, People's Hospital of Longhua Shenzhen, Shenzhen, 518109, Guangdong, China.
| | - Bin Wu
- Department of Stomatology, People's Hospital of Longhua Shenzhen, Shenzhen, 518109, Guangdong, China
| | - Weidong Tian
- State Key Laboratory of Oral Disease & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Oral Regenerative Medicine, West China School of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
10
|
Alternative Methods as Tools for Obesity Research: In Vitro and In Silico Approaches. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010108. [PMID: 36676057 PMCID: PMC9860640 DOI: 10.3390/life13010108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023]
Abstract
The study of adipogenesis is essential for understanding and treating obesity, a multifactorial problem related to body fat accumulation that leads to several life-threatening diseases, becoming one of the most critical public health problems worldwide. In this review, we propose to provide the highlights of the adipogenesis study based on in vitro differentiation of human mesenchymal stem cells (hMSCs). We list in silico methods, such as molecular docking for identification of molecular targets, and in vitro approaches, from 2D, more straightforward and applied for screening large libraries of substances, to more representative physiological models, such as 3D and bioprinting models. We also describe the development of physiological models based on microfluidic systems applied to investigate adipogenesis in vitro. We intend to identify the main alternative models for adipogenesis evaluation, contributing to the direction of preclinical research in obesity. Future directions indicate the association of in silico and in vitro techniques to bring a clear picture of alternative methods based on adipogenesis as a tool for obesity research.
Collapse
|
11
|
Amine ZE, Mauger JF, Imbeault P. CYP1A1, VEGFA and Adipokine Responses of Human Adipocytes Co-exposed to PCB126 and Hypoxia. Cells 2022; 11:cells11152282. [PMID: 35892579 PMCID: PMC9331964 DOI: 10.3390/cells11152282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022] Open
Abstract
It is increasingly recognized that hypoxia may develop in adipose tissue as its mass expands. Adipose tissue is also the main reservoir of lipophilic pollutants, including polychlorinated biphenyls (PCBs). Both hypoxia and PCBs have been shown to alter adipose tissue functions. The signaling pathways induced by hypoxia and pollutants may crosstalk, as they share a common transcription factor: aryl hydrocarbon receptor nuclear translocator (ARNT). Whether hypoxia and PCBs crosstalk and affect adipokine secretion in human adipocytes remains to be explored. Using primary human adipocytes acutely co-exposed to different levels of hypoxia (24 h) and PCB126 (48 h), we observed that hypoxia significantly inhibits the PCB126 induction of cytochrome P450 (CYP1A1) transcription in a dose-response manner, and that Acriflavine (ACF)—an HIF1α inhibitor—partially restores the PCB126 induction of CYP1A1 under hypoxia. On the other hand, exposure to PCB126 did not affect the transcription of the vascular endothelial growth factor-A (VEGFA) under hypoxia. Exposure to hypoxia increased leptin and interleukin-6 (IL-6), and decreased adiponectin levels dose-dependently, while PCB126 increased IL-6 and IL-8 secretion in a dose-dependent manner. Co-exposure to PCB126 and hypoxia did not alter the adipokine secretion pattern observed under hypoxia and PCB126 exposure alone. In conclusion, our results indicate that (1) hypoxia inhibits PCB126-induced CYP1A1 expression at least partly through ARNT-dependent means, suggesting that hypoxia could affect PCB metabolism and toxicity in adipose tissue, and (2) hypoxia and PCB126 affect leptin, adiponectin, IL-6 and IL-8 secretion differently, with no apparent crosstalk between the two factors.
Collapse
Affiliation(s)
- Zeinab El Amine
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (Z.E.A.); (J.-F.M.)
| | - Jean-François Mauger
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (Z.E.A.); (J.-F.M.)
| | - Pascal Imbeault
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (Z.E.A.); (J.-F.M.)
- Institut du Savoir Montfort, Hôpital Montfort, Ottawa, ON K1K 0T2, Canada
- Correspondence: ; Tel.: +1-(613)-562-5800-(7290)
| |
Collapse
|
12
|
Barisón MJ, Nogoceke R, Josino R, Horinouchi CDDS, Marcon BH, Correa A, Stimamiglio MA, Robert AW. Functionalized Hydrogels for Cartilage Repair: The Value of Secretome-Instructive Signaling. Int J Mol Sci 2022; 23:ijms23116010. [PMID: 35682690 PMCID: PMC9181449 DOI: 10.3390/ijms23116010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/07/2023] Open
Abstract
Cartilage repair has been a challenge in the medical field for many years. Although treatments that alleviate pain and injury are available, none can effectively regenerate the cartilage. Currently, regenerative medicine and tissue engineering are among the developed strategies to treat cartilage injury. The use of stem cells, associated or not with scaffolds, has shown potential in cartilage regeneration. However, it is currently known that the effect of stem cells occurs mainly through the secretion of paracrine factors that act on local cells. In this review, we will address the use of the secretome—a set of bioactive factors (soluble factors and extracellular vesicles) secreted by the cells—of mesenchymal stem cells as a treatment for cartilage regeneration. We will also discuss methodologies for priming the secretome to enhance the chondroregenerative potential. In addition, considering the difficulty of delivering therapies to the injured cartilage site, we will address works that use hydrogels functionalized with growth factors and secretome components. We aim to show that secretome-functionalized hydrogels can be an exciting approach to cell-free cartilage repair therapy.
Collapse
|
13
|
Hypoxia-Preconditioned Extracellular Vesicles from Mesenchymal Stem Cells Improve Cartilage Repair in Osteoarthritis. MEMBRANES 2022; 12:membranes12020225. [PMID: 35207146 PMCID: PMC8875566 DOI: 10.3390/membranes12020225] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/24/2021] [Accepted: 02/06/2022] [Indexed: 02/07/2023]
Abstract
In the past decade, mesenchymal stem cells (MSCs) have been widely used for the treatment of osteoarthritis (OA), and extracellular vesicles (EVs) may play a major role in the efficacy of this treatment. Hypoxia can change the cargo and biological functions of MSC-derived EVs (MSC-EVs). The aim of the present study was to determine whether the effects of hypoxia-preconditioned MSC-EVs on OA cartilage repair are superior to normoxia-preconditioned MSC-EVs. By using in vitro and in vivo OA models, we verified that hypoxia-preconditioned MSC-EVs improved chondrocyte proliferation and migration and suppressed chondrocyte apoptosis to a greater extent than normoxia-preconditioned MSC-EVs. Furthermore, we found that hypoxia altered the microRNA expression in MSC-EVs and identified four differentially expressed microRNAs: hsa-miR-181c-5p, hsa-miR-18a-3p, hsa-miR-376a-5p, and hsa-miR-337-5p. Bioinformatics analysis revealed that hypoxic pretreatment may promote cartilage repair by stimulating chondrocyte proliferation and migration and suppressing chondrocyte apoptosis through the miRNA-18-3P/JAK/STAT or miRNA-181c-5p/MAPK signaling pathway. Therefore, hypoxia-preconditioned EVs may be a novel treatment for OA.
Collapse
|
14
|
Milan G, Conci S, Sanna M, Favaretto F, Bettini S, Vettor R. ASCs and their role in obesity and metabolic diseases. Trends Endocrinol Metab 2021; 32:994-1006. [PMID: 34625375 DOI: 10.1016/j.tem.2021.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/23/2021] [Accepted: 09/03/2021] [Indexed: 01/04/2023]
Abstract
We describe adipose stromal/stem cells (ASCs) in the structural/functional context of the adipose tissue (AT) stem niche (adiponiche), including cell-cell interactions and the microenvironment, and emphasize findings obtained in humans and in lineage-tracing models. ASCs have distinctive markers, 'colors', and anatomical 'locations' which influence their functions. Each adiponiche component can become impaired, thereby contributing to the pathological AT alterations seen in obesity and metabolic diseases. We discuss adiposopathy with a focus on adiponiche dysfunction, and underline the mechanisms that control AT expansion and energy balance. Better understanding of adiponiche regulation and ASC features could help to identify therapeutic targets that favor weight loss and counteract weight regain, and also contribute to innovative strategies for regenerative medicine.
Collapse
Affiliation(s)
- Gabriella Milan
- Department of Medicine, University of Padua, Internal Medicine 3, 35128 Padua, Italy; Center for the Study and the Integrated Treatment of Obesity, Padua Hospital, 35128 Padua, Italy.
| | - Scilla Conci
- Department of Medicine, University of Padua, Internal Medicine 3, 35128 Padua, Italy; Center for the Study and the Integrated Treatment of Obesity, Padua Hospital, 35128 Padua, Italy
| | - Marta Sanna
- Department of Medicine, University of Padua, Internal Medicine 3, 35128 Padua, Italy; Center for the Study and the Integrated Treatment of Obesity, Padua Hospital, 35128 Padua, Italy
| | - Francesca Favaretto
- Department of Medicine, University of Padua, Internal Medicine 3, 35128 Padua, Italy; Center for the Study and the Integrated Treatment of Obesity, Padua Hospital, 35128 Padua, Italy
| | - Silvia Bettini
- Department of Medicine, University of Padua, Internal Medicine 3, 35128 Padua, Italy; Center for the Study and the Integrated Treatment of Obesity, Padua Hospital, 35128 Padua, Italy
| | - Roberto Vettor
- Department of Medicine, University of Padua, Internal Medicine 3, 35128 Padua, Italy; Center for the Study and the Integrated Treatment of Obesity, Padua Hospital, 35128 Padua, Italy
| |
Collapse
|
15
|
Effect of biomolecules derived from human platelet-rich plasma on the ex vivo expansion of human adipose-derived mesenchymal stem cells for clinical applications. Biologicals 2021; 75:37-48. [PMID: 34785135 DOI: 10.1016/j.biologicals.2021.11.001] [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: 07/20/2020] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 11/20/2022] Open
Abstract
Mesenchymal stem cells are a tool in cell therapies but demand a large cell number per treatment, for that, suitable culture media is required which contains fetal bovine serum (FBS). However, for cell-based therapy applications, the use of FBS is problematic. Several alternatives to FBS have been explored, including human derivatives from platelet-rich plasma (hD-PRP). Although various studies have evaluated the impact of hD-PRP on MSC proliferation and differentiation, few of them have assessed their influence on processes, such as metabolism and gene expression. Here, we cultured human adipose-derived MSCs (hAD-MSCs) in media supplemented with either 10% hD-PRP (hD-PRP-SM) or 10% FBS (FBS-SM) in order to characterize them and evaluate the effect of hD-PRP on cell metabolism, gene expression of associated regenerative factors, as well as chromosome stability during cell expansion. We found that hAD-MSCs cultured in hD-PRP-SM have a greater cell elongation but express similar surface markers; in addition, hD-PRP-SM promoted a significant osteogenic differentiation in the absence of differentiation medium and increased the growth rate, maintaining chromosomal stability. In terms of cell metabolic profile, hAD-MSC behavior did not reveal any differences between both culture conditions. Conversely, significant differences in collagen I and angiopoietin 2 expression were observed between both conditions. The present results suggest that hD-PRP may influence hAD-MSC behavior.
Collapse
|
16
|
Yang Y, Lee EH, Yang Z. Hypoxia conditioned mesenchymal stem cells in tissue regeneration application. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:966-977. [PMID: 34569290 DOI: 10.1089/ten.teb.2021.0145] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) have been demonstrated as promising cell sources for tissue regeneration due to their capability of self-regeneration, differentiation and immunomodulation. MSCs also exert extensive paracrine effects through release of trophic factors and extracellular vesicles. However, despite extended exploration of MSCs in pre-clinical studies, the results are far from satisfactory due to the poor engraftment and low level of survival after implantation. Hypoxia preconditioning has been proposed as an engineering approach to improve the therapeutic potential of MSCs. During in vitro culture, hypoxic conditions can promote MSC proliferation, survival and migration through various cellular responses to the reduction of oxygen tension. The multilineage differentiation potential of MSCs is altered under hypoxia, with consistent reports of enhanced chondrogenesis. Hypoxia also stimulates the paracrine activities of MSCs and increases the production of secretome both in terms of soluble factors as well as extracellular vesicles. The secretome from hypoxia preconditioned MSCs play important roles in promoting cell proliferation and migration, enhancing angiogenesis while inhibiting apoptosis and inflammation. In this review, we summarise current knowledge of hypoxia-induced changes in MSCs and discuss the application of hypoxia preconditioned MSCs as well as hypoxic secretome in different kinds of disease models.
Collapse
Affiliation(s)
- Yanmeng Yang
- National University of Singapore, 37580, Orthopaedic Surgery, 27 Medical Drive, Singapore, Singapore, 117510;
| | - Eng Hin Lee
- National University of Singapore, Department of Orthopaedic Surgery, 1E Kent Ridge Road, NUHS Tower Block, Level 11, Singapore, Singapore, 119228;
| | - Zheng Yang
- National University of Singapore, Life Sciences Institute, Singapore, Singapore;
| |
Collapse
|
17
|
Silvestris N, Argentiero A, Natalicchio A, D'Oronzo S, Beretta GD, Acquati S, Adinolfi V, Di Bartolo P, Danesi R, Faggiano A, Ferrari P, Gallo M, Gori S, Morviducci L, Russo A, Tuveri E, Zatelli MC, Montagnani M, Giorgino F. Antineoplastic dosing in overweight and obese cancer patients: an Associazione Italiana Oncologia Medica (AIOM)/Associazione Medici Diabetologi (AMD)/Società Italiana Endocrinologia (SIE)/Società Italiana Farmacologia (SIF) multidisciplinary consensus position paper. ESMO Open 2021; 6:100153. [PMID: 33984679 PMCID: PMC8134762 DOI: 10.1016/j.esmoop.2021.100153] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/23/2021] [Accepted: 04/14/2021] [Indexed: 01/22/2023] Open
Abstract
Most anticancer molecules are administered in body-size-based dosing schedules, bringing up unsolved issues regarding pharmacokinetic data in heavy patients. The worldwide spread of obesity has not been matched by improved methods and strategies for tailored drug dosage in this population. The weight or body surface area (BSA)-based approaches may fail to fully reflect the complexity of the anthropometric features besides obesity in cancer patients suffering from sarcopenia. Likewise, there is a lack of pharmacokinetic data on obese patients for the majority of chemotherapeutic agents as well as for new target drugs and immunotherapy. Therefore, although the available findings point to the role of dose intensity in cancer treatment, and support full weight-based dosing, empirical dose capping often occurs in clinical practice in order to avoid toxicity. Thus a panel of experts of the Associazione Italiana Oncologia Medica (AIOM), Associazione Medici Diabetologi (AMD), Società Italiana Endocrinologia (SIE), and Società Italiana Farmacologia (SIF), provides here a consensus statement for appropriate cytotoxic chemotherapy and new biological cancer drug dosing in obese patients. The worldwide spread of obesity is an emerging challenge also in cancer patients Weight or BSA-based approaches do not adequately address the critical issue of optimal dosing for cancer drugs under obesity Empirical dose capping is often employed in clinical practice to avoid toxicities among overweight and obese patients There is a lack of clinical and pharmacokinetic studies in this population Clinical practice recommendations should guide suitable dosing of cytotoxic and biological cancer drugs in obese patients
Collapse
Affiliation(s)
- N Silvestris
- Medical Oncology Unit, IRCCS Istituto Tumori 'Giovanni Paolo II', Bari, Italy; Department of Biomedical Sciences and Human Oncology, Division of Medical Oncology, University of Bari Aldo Moro, Bari, Italy.
| | - A Argentiero
- Medical Oncology Unit, IRCCS Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - A Natalicchio
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - S D'Oronzo
- Department of Biomedical Sciences and Human Oncology, Division of Medical Oncology, University of Bari Aldo Moro, Bari, Italy; Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
| | - G D Beretta
- Medical Oncology Department, Humanitas Gavazzeni, Bergamo, Italy
| | - S Acquati
- Endocrinology Unit, Ospedale Pierantoni-Morgagni, Forlì, Italy
| | - V Adinolfi
- Endocrinology and Diabetology Unit, ASL Verbano Cusio Ossola, Domodossola, Italy
| | - P Di Bartolo
- Diabetology Clinic, Rete Clinica di Diabetologia Aziendale - Dipartimento, Internistico di Ravenna - AUSL Romagna, Ravenna, Italy
| | - R Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - A Faggiano
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - P Ferrari
- Palliative Care Unit, Istituti Clinici Scientifici Maugeri SPA SB, IRCCS (PV), Pavia PV, Italy
| | - M Gallo
- Endocrinology and Metabolic Diseases Unit of AO SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - S Gori
- Oncologia Medica, IRCCS Ospedale Don Calabria-Sacro Cuore di Negrar, Verona, Italy
| | - L Morviducci
- Diabetology and Nutrition Unit, Department of Medical Specialities, ASL Roma 1 - S. Spirito Hospital, Rome, Italy
| | - A Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - E Tuveri
- Diabetology, Endocrinology and Metabolic Diseases Service, ATS Sardegna - ASSL Carbonia-Iglesias, Italy
| | - M C Zatelli
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - M Montagnani
- Department of Biomedical Sciences and Human Oncology, Division of Medical Oncology, University of Bari Aldo Moro, Bari, Italy
| | - F Giorgino
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
18
|
Han TTY, Flynn LE. Perfusion bioreactor culture of human adipose‐derived stromal cells on decellularized adipose tissue scaffolds enhances in vivo adipose tissue regeneration. J Tissue Eng Regen Med 2020; 14:1827-1840. [DOI: 10.1002/term.3133] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/02/2020] [Accepted: 09/17/2020] [Indexed: 12/22/2022]
Affiliation(s)
- Tim Tian Y. Han
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry The University of Western Ontario London Ontario Canada
| | - Lauren E. Flynn
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry The University of Western Ontario London Ontario Canada
- Department of Chemical and Biochemical Engineering, Thompson Engineering Building The University of Western Ontario London Ontario Canada
- Bone and Joint Institute The University of Western Ontario London Ontario Canada
| |
Collapse
|
19
|
Peng SL, Chen CM. The influence of obesity on cerebral blood flow in young adults using arterial spin labeling MRI. NMR IN BIOMEDICINE 2020; 33:e4375. [PMID: 32729160 DOI: 10.1002/nbm.4375] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 06/11/2023]
Abstract
Obesity causes damage to several organs, including the brain. Recent studies have been focusing on understanding the mechanisms through which obesity affects brain structure and function using neuroimaging techniques. A functional biomarker, such as cerebral blood flow (CBF), is a powerful tool that can be used to explore neural dysfunction. However, there is currently limited information regarding the association between CBF and obesity. The study was conducted to investigate the potential effect of obesity on brain perfusion in a young cohort aged 20-30 years. A total of 21 obese (body mass index (BMI) > 26 kg/m2 ) and 21 lean (BMI < 24 kg/m2 ) right-handed volunteers were included in this study. CBF was acquired using the 2D single post-labeling delay (PLD) arterial spin labeling (ASL) technique on a 3 T MRI scanner. A multiple regression analysis was performed to examine the difference in global and regional gray matter (GM) CBF between the groups. CBF value was assigned as the dependent variable, whereas age, sex, and group (obese or lean) were considered as the independent variables. Results showed that group-related differences in CBF were homogeneous across brain regions, as obese subjects had significantly lower global GM CBF than lean subjects (P < 0.05). In the voxelwise analysis, obese individuals had significantly lower CBF in the left pulvinar of the thalamus and visual association areas, including Brodmann area (BA) 7, BA18, and BA19, than lean subjects. Although the signal-to-noise ratio was slightly compromised for 2D sequences and subject-specific arterial transit time was not estimated due to a single PLD sequence, this study demonstrated alterations in CBF in obese subjects, particularly in regions of the pulvinar of the thalamus and its synchronously related areas such as visual association areas. These results suggest that ASL provides a potential platform for further obesity-related research.
Collapse
Affiliation(s)
- Shin-Lei Peng
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Chun-Ming Chen
- Department of Radiology, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
20
|
Swartz HM, Flood AB, Schaner PE, Halpern H, Williams BB, Pogue BW, Gallez B, Vaupel P. How best to interpret measures of levels of oxygen in tissues to make them effective clinical tools for care of patients with cancer and other oxygen-dependent pathologies. Physiol Rep 2020; 8:e14541. [PMID: 32786045 PMCID: PMC7422807 DOI: 10.14814/phy2.14541] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023] Open
Abstract
It is well understood that the level of molecular oxygen (O2 ) in tissue is a very important factor impacting both physiology and pathological processes as well as responsiveness to some treatments. Data on O2 in tissue could be effectively utilized to enhance precision medicine. However, the nature of the data that can be obtained using existing clinically applicable techniques is often misunderstood, and this can confound the effective use of the information. Attempts to make clinical measurements of O2 in tissues will inevitably provide data that are aggregated over time and space and therefore will not fully represent the inherent heterogeneity of O2 in tissues. Additionally, the nature of existing techniques to measure O2 may result in uneven sampling of the volume of interest and therefore may not provide accurate information on the "average" O2 in the measured volume. By recognizing the potential limitations of the O2 measurements, one can focus on the important and useful information that can be obtained from these techniques. The most valuable clinical characterizations of oxygen are likely to be derived from a series of measurements that provide data about factors that can change levels of O2 , which then can be exploited both diagnostically and therapeutically. The clinical utility of such data ultimately needs to be verified by careful studies of outcomes related to the measured changes in levels of O2 .
Collapse
Affiliation(s)
- Harold M Swartz
- Department of Radiology, Dartmouth Medical School, Hanover, NH, USA
- Department of Medicine, Section of Radiation Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Ann Barry Flood
- Department of Radiology, Dartmouth Medical School, Hanover, NH, USA
| | - Philip E Schaner
- Department of Medicine, Section of Radiation Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Howard Halpern
- Department Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Benjamin B Williams
- Department of Radiology, Dartmouth Medical School, Hanover, NH, USA
- Department of Medicine, Section of Radiation Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Brian W Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Bernard Gallez
- Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Peter Vaupel
- Department Radiation Oncology, University Medical Center, University of Freiburg, Freiburg, Germany
- German Cancer Center Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
21
|
Lempesis IG, Meijel RLJ, Manolopoulos KN, Goossens GH. Oxygenation of adipose tissue: A human perspective. Acta Physiol (Oxf) 2020; 228:e13298. [PMID: 31077538 PMCID: PMC6916558 DOI: 10.1111/apha.13298] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/03/2019] [Accepted: 05/08/2019] [Indexed: 12/13/2022]
Abstract
Obesity is a complex disorder of excessive adiposity, and is associated with adverse health effects such as cardiometabolic complications, which are to a large extent attributable to dysfunctional white adipose tissue. Adipose tissue dysfunction is characterized by adipocyte hypertrophy, impaired adipokine secretion, a chronic low‐grade inflammatory status, hormonal resistance and altered metabolic responses, together contributing to insulin resistance and related chronic diseases. Adipose tissue hypoxia, defined as a relative oxygen deficit, in obesity has been proposed as a potential contributor to adipose tissue dysfunction, but studies in humans have yielded conflicting results. Here, we will review the role of adipose tissue oxygenation in the pathophysiology of obesity‐related complications, with a specific focus on human studies. We will provide an overview of the determinants of adipose tissue oxygenation, as well as the role of adipose tissue oxygenation in glucose homeostasis, lipid metabolism and inflammation. Finally, we will discuss the putative effects of physiological and experimental hypoxia on adipose tissue biology and whole‐body metabolism in humans. We conclude that several lines of evidence suggest that alteration of adipose tissue oxygenation may impact metabolic homeostasis, thereby providing a novel strategy to combat chronic metabolic diseases in obese humans.
Collapse
Affiliation(s)
- Ioannis G. Lempesis
- College of Medical and Dental Sciences, Institute of Metabolism and Systems Research (IMSR) University of Birmingham Birmingham UK
- Centre for Endocrinology, Diabetes and Metabolism Birmingham Health Partners Birmingham UK
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism Maastricht University Medical Centre Maastricht the Netherlands
| | - Rens L. J. Meijel
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism Maastricht University Medical Centre Maastricht the Netherlands
| | - Konstantinos N. Manolopoulos
- College of Medical and Dental Sciences, Institute of Metabolism and Systems Research (IMSR) University of Birmingham Birmingham UK
- Centre for Endocrinology, Diabetes and Metabolism Birmingham Health Partners Birmingham UK
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism Maastricht University Medical Centre Maastricht the Netherlands
| |
Collapse
|
22
|
Zhu Q, Shan C, Li L, Song L, Zhang K, Zhou Y. Differential expression of genes associated with hypoxia pathway on bone marrow stem cells in osteoporosis patients with different bone mass index. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:309. [PMID: 31475179 DOI: 10.21037/atm.2019.06.27] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background This study aimed to assess gene expression changes associated with hypoxia pathway on bone marrow stem cells (BMSCs) and explore effects of bone mass index (BMI) on hypoxia pathway of osteoporosis (OP) patients. Methods Human BMSCs were isolated from bone marrow. Subjects were divided into healthy control group and OP group which was further divided into BMI <25 OP subgroup and BMI ≥25 OP subgroup. Results The genes downregulated in OP patients were involved in hypoxia pathway. Furthermore, those genes were even downregulated in OP patients BMI ≥25 subgroup than OP patients BMI <25 subgroup. The genes were expressed in response to decreased oxygen levels, and their functions are related to photoperiodism, positive regulation of myoblast differentiation, and entrainment of circadian clock by gene ontology (GO) analysis. Conclusions The expression of genes associated with hypoxia pathway on BMSCs in OP patients are lower than healthy subjects, and the expression of genes related to carbohydrate metabolism are lower in overweight OP patients than in normal weight OP patients. These results need further research.
Collapse
Affiliation(s)
- Qi Zhu
- The Endocrinology Department of Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,The Geriatric Department of Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Chang Shan
- The Endocrinology Department of Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,Department of Endocrine and Metabolic Diseases, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai 200025, China
| | - Ling Li
- The Endocrinology Department of Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Lige Song
- The Endocrinology Department of Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Keqin Zhang
- The Endocrinology Department of Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Yun Zhou
- The Endocrinology Department of Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| |
Collapse
|
23
|
Norouzirad R, Gholami H, Ghanbari M, Hedayati M, González-Muniesa P, Jeddi S, Ghasemi A. Dietary inorganic nitrate attenuates hyperoxia-induced oxidative stress in obese type 2 diabetic male rats. Life Sci 2019; 230:188-196. [PMID: 31150686 DOI: 10.1016/j.lfs.2019.05.068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/18/2019] [Accepted: 05/26/2019] [Indexed: 10/26/2022]
Abstract
AIMS Hyperoxia has beneficial metabolic effects in type 2 diabetes. However, hyperoxia exacerbates already existing oxidative stress in type 2 diabetes. Nitrate, a nitric oxide donor, is an effective new treatment in type 2 diabetes and also has antioxidant properties. The aim of this study was to determine whether nitrate administration can attenuate hyperoxia-induced oxidative stress in obese type 2 diabetic rats. MAIN METHODS Fifty-six male Wistar rats (190-210 g) were divided into 8 groups: Controls (non-treated, nitrate-treated, O2-treated, and nitrate + O2-treated) and diabetes (non-treated, nitrate-treated, O2-treated, and nitrate + O2-treated). Diabetes was induced using high-fat diet and low-dose of streptozotocin (30 mg/kg). Rats in intervention groups, were exposed to 95% oxygen and consumed sodium nitrate (100 mg/L) in drinking water. Serum fasting glucose, oxidized (GSSG) and reduced (GSH) glutathiones, total oxidant status (TOS), catalase and superoxide dismutase (SOD) activities, and total antioxidant capacity (TAC) were measured after intervention. Oxidative stress index (OSI) was calculated as TOS/TAC ratio. KEY FINDINGS Diabetic rats had increased oxidative stress and hyperoxia exacerbated it. In O2-diabetic rats, nitrate decreased GSSG (102.7 ± 2.1 vs. 236.0 ± 20.1 μM, P < 0.001), TOS (67.7 ± 7.3 vs. 104 ± 3.8 μM, P < 0.001), and OSI (0.44 ± 0.04 vs. 0.91 ± 0.07, P < 0.001) and increased catalase (2.8 ± 0.13 vs. 1.8 ± 0.21 KU/L, P = 0.014), SOD (53.4 ± 1.5 vs. 38.4 ± 1.2 U/mL, P < 0.001), GSH (43.7 ± 1.4 vs. 17.8 ± 0.5 mM, P = 0.003), TAC (152.5 ± 1.9 vs. 116.7 ± 5.0 mM, P < 0.001), and GSH/GSSG ratio (0.43 ± 0.01 vs. 0.08 ± 0.01, P = 0.005). Nitrate also potentiated effects of hyperoxia on decreasing fasting glucose. SIGNIFICANCE Our results showed that dietary nitrate attenuates hyperoxia-induced oxidative stress in type 2 diabetic rats.
Collapse
Affiliation(s)
- Reza Norouzirad
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Dezful University of Medical Sciences, Dezful, Iran.
| | - Hanieh Gholami
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Ghanbari
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Pedro González-Muniesa
- University of Navarra, Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, Pamplona, Spain; University of Navarra, Centre for Nutrition Research, School of Pharmacy and Nutrition, Pamplona, Spain; IdiSNA Navarra's Health Research Institute, Pamplona, Spain; CIBERobn Physiopathology of Obesity and Nutrition, Centre of Biomedical Research Network, ISCIII, Madrid, Spain.
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Rodriguez LA, Mohammadipoor A, Alvarado L, Kamucheka RM, Asher AM, Cancio LC, Antebi B. Preconditioning in an Inflammatory Milieu Augments the Immunotherapeutic Function of Mesenchymal Stromal Cells. Cells 2019; 8:cells8050462. [PMID: 31096722 PMCID: PMC6562603 DOI: 10.3390/cells8050462] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/09/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) have emerged as potent therapeutic agents for multiple indications. However, recent evidence indicates that MSC function is compromised in the physiological post-injury milieu. In this study, bone marrow (BM)- and adipose-derived (AD)-MSCs were preconditioned in hypoxia with or without inflammatory mediators to potentiate their immunotherapeutic function in preparation for in vivo delivery. Human MSCs were cultured for 48 hours in either normoxia (21% O2) or hypoxia (2% O2) with or without the addition of Cytomix, thus creating 4 groups: 1) normoxia (21%); 2) Cytomix-normoxia (+21%); 3) hypoxia (2%); and 4) Cytomix-hypoxia (+2%). The 4 MSC groups were subjected to comprehensive evaluation of their characteristics and function. Preconditioning did not alter common MSC surface markers; nonetheless, Cytomix treatment triggered an increase in tissue factor (TF) expression. Moreover, the BM-MSCs and AD-MSCs from the +2% group were not able to differentiate to chondrocytes and osteoblasts, respectively. Following Cytomix preconditioning, the metabolism of MSCs was significantly increased while viability was decreased in AD-MSCs, but not in BM-MSCs. MSCs from both tissues showed a significant upregulation of key anti-inflammatory genes, increased secretion of IL-1 receptor antagonist (RA), and enhanced suppression of T-cell proliferation following the Cytomix treatment. Similarly, following a lipopolysaccharide challenge, the Cytomix-treated MSCs suppressed TNF-α secretion, while promoting the production of IL-10 and IL-1RA. These preconditioning approaches facilitate the production of MSCs with robust anti-inflammatory properties. AD-MSCs preconditioned with Cytomix under normoxia appear to be the most promising therapeutic candidates; however, safety concerns, such as thrombogenic disposition of cells due to TF expression, should be carefully considered prior to clinical translation.
Collapse
Affiliation(s)
- Luis A Rodriguez
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.
| | - Arezoo Mohammadipoor
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA.
| | - Lucero Alvarado
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA.
- University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - Robin M Kamucheka
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.
| | - Amber M Asher
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA.
| | - Leopoldo C Cancio
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.
| | - Ben Antebi
- United States Army Institute of Surgical Research, San Antonio, TX 78234, USA.
- University of Texas at San Antonio, San Antonio, TX 78249, USA.
| |
Collapse
|
25
|
Norouzirad R, Ghanbari M, Bahadoran Z, Abdollahifar MA, Rasouli N, Ghasemi A. Hyperoxia improves carbohydrate metabolism by browning of white adipocytes in obese type 2 diabetic rats. Life Sci 2019; 220:58-68. [DOI: 10.1016/j.lfs.2019.01.045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/20/2019] [Accepted: 01/27/2019] [Indexed: 02/07/2023]
|
26
|
Insulin Controls Triacylglycerol Synthesis through Control of Glycerol Metabolism and Despite Increased Lipogenesis. Nutrients 2019; 11:nu11030513. [PMID: 30823376 PMCID: PMC6470968 DOI: 10.3390/nu11030513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 02/22/2019] [Accepted: 02/22/2019] [Indexed: 12/12/2022] Open
Abstract
Under normoxic conditions, adipocytes in primary culture convert huge amounts of glucose to lactate and glycerol. This “wasting” of glucose may help to diminish hyperglycemia. Given the importance of insulin in the metabolism, we have studied how it affects adipocyte response to varying glucose levels, and whether the high basal conversion of glucose to 3-carbon fragments is affected by insulin. Rat fat cells were incubated for 24 h in the presence or absence of 175 nM insulin and 3.5, 7, or 14 mM glucose; half of the wells contained 14C-glucose. We analyzed glucose label fate, medium metabolites, and the expression of key genes controlling glucose and lipid metabolism. Insulin increased both glucose uptake and the flow of carbon through glycolysis and lipogenesis. Lactate excretion was related to medium glucose levels, which agrees with the purported role of disposing excess (circulating) glucose. When medium glucose was low, most basal glycerol came from lipolysis, but when glucose was high, release of glycerol via breakup of glycerol-3P was predominant. Although insulin promotes lipogenesis, it also limited the synthesis of glycerol-3P from glucose and its incorporation into acyl-glycerols. We assume that this is a mechanism of adipose tissue defense to avoid crippling fat accumulation which has not yet been described.
Collapse
|
27
|
Keeley TP, Mann GE. Defining Physiological Normoxia for Improved Translation of Cell Physiology to Animal Models and Humans. Physiol Rev 2019; 99:161-234. [PMID: 30354965 DOI: 10.1152/physrev.00041.2017] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The extensive oxygen gradient between the air we breathe (Po2 ~21 kPa) and its ultimate distribution within mitochondria (as low as ~0.5-1 kPa) is testament to the efforts expended in limiting its inherent toxicity. It has long been recognized that cell culture undertaken under room air conditions falls short of replicating this protection in vitro. Despite this, difficulty in accurately determining the appropriate O2 levels in which to culture cells, coupled with a lack of the technology to replicate and maintain a physiological O2 environment in vitro, has hindered addressing this issue thus far. In this review, we aim to address the current understanding of tissue Po2 distribution in vivo and summarize the attempts made to replicate these conditions in vitro. The state-of-the-art techniques employed to accurately determine O2 levels, as well as the issues associated with reproducing physiological O2 levels in vitro, are also critically reviewed. We aim to provide the framework for researchers to undertake cell culture under O2 levels relevant to specific tissues and organs. We envisage that this review will facilitate a paradigm shift, enabling translation of findings under physiological conditions in vitro to disease pathology and the design of novel therapeutics.
Collapse
Affiliation(s)
- Thomas P Keeley
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London , London , United Kingdom
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London , London , United Kingdom
| |
Collapse
|
28
|
van Meijel RL, Blaak EE, Goossens GH. Adipose tissue metabolism and inflammation in obesity. MECHANISMS AND MANIFESTATIONS OF OBESITY IN LUNG DISEASE 2019:1-22. [DOI: 10.1016/b978-0-12-813553-2.00001-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
29
|
Cucchi F, Rossmeislova L, Simonsen L, Jensen MR, Bülow J. A vicious circle in chronic lymphoedema pathophysiology? An adipocentric view. Obes Rev 2017; 18:1159-1169. [PMID: 28660651 DOI: 10.1111/obr.12565] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/07/2017] [Accepted: 04/25/2017] [Indexed: 12/11/2022]
Abstract
Chronic lymphoedema is a disease caused by a congenital or acquired damage to the lymphatic system and characterized by complex chains of pathophysiologic events such as lymphatic fluid stasis, chronic inflammation, lymphatic vessels impairment, adipose tissue deposition and fibrosis. These events seem to maintain and reinforce themselves through a positive feedback loop: regardless of the initial cause of lymphatic stasis, the dysfunctional adipose tissue and its secretion products can worsen lymphatic vessels' function, aggravating lymph leakage and stagnation, which can promote further adipose tissue deposition and fibrosis, similar to what may happen in obesity. In addition to the current knowledge about the tight and ancestral interrelation between immunity system and metabolism, there is evidence for similarities between obesity-related and lymphatic damage-induced lymphoedema. Together, these observations indicate strong reciprocal relationship between lymphatics and adipose tissue and suggest a possible key role of the adipocyte in the pathophysiology of chronic lymphoedema's vicious circle.
Collapse
Affiliation(s)
- F Cucchi
- Department of Clinical Physiology and Nuclear Medicine, Bispebjerg and Frederiksberg Hospitals, Copenhagen, Denmark
| | - L Rossmeislova
- Department for the Study of Obesity and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - L Simonsen
- Department of Clinical Physiology and Nuclear Medicine, Bispebjerg and Frederiksberg Hospitals, Copenhagen, Denmark
| | - M R Jensen
- Department of Clinical Physiology and Nuclear Medicine, Bispebjerg and Frederiksberg Hospitals, Copenhagen, Denmark
| | - J Bülow
- Department of Clinical Physiology and Nuclear Medicine, Bispebjerg and Frederiksberg Hospitals, Copenhagen, Denmark.,Department of Biomedical Sciences, Copenhagen University, Denmark
| |
Collapse
|
30
|
Franconi F, Lemaire L, Saint‐Jalmes H, Saulnier P. Tissue oxygenation mapping by combined chemical shift and T
1
magnetic resonance imaging. Magn Reson Med 2017; 79:1981-1991. [DOI: 10.1002/mrm.26857] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/22/2017] [Accepted: 07/07/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Florence Franconi
- PRISM Plate‐forme de recherche en imagerie et spectroscopie multi‐modales, PRISM‐Icat, Angers et PRISM‐Biosit CNRS UMS 3480, INSERM UMS 018, Rennes, UBL Universite BretagneLoire France
- Micro & Nanomédecines Translationelles‐MINT, UNIV Angers, INSERM U1066, CNRS UMR 6021UBL Universite Bretagne LoireAngers France
| | - Laurent Lemaire
- PRISM Plate‐forme de recherche en imagerie et spectroscopie multi‐modales, PRISM‐Icat, Angers et PRISM‐Biosit CNRS UMS 3480, INSERM UMS 018, Rennes, UBL Universite BretagneLoire France
- Micro & Nanomédecines Translationelles‐MINT, UNIV Angers, INSERM U1066, CNRS UMR 6021UBL Universite Bretagne LoireAngers France
| | - Hervé Saint‐Jalmes
- PRISM Plate‐forme de recherche en imagerie et spectroscopie multi‐modales, PRISM‐Icat, Angers et PRISM‐Biosit CNRS UMS 3480, INSERM UMS 018, Rennes, UBL Universite BretagneLoire France
- INSERM, UMR 1099Rennes France
- LTSI, Université de Rennes 1Rennes France
- CRLCC, Centre Eugène MarquisRennes France
| | - Patrick Saulnier
- Micro & Nanomédecines Translationelles‐MINT, UNIV Angers, INSERM U1066, CNRS UMR 6021UBL Universite Bretagne LoireAngers France
| |
Collapse
|
31
|
Rumpler M, Hajnsek M, Baumann P, Pieber TR, Klimant I. Monitoring tissue oxygen heterogeneities and their influence on optical glucose measurements in an animal model. J Clin Monit Comput 2017; 32:583-586. [DOI: 10.1007/s10877-017-0034-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/30/2017] [Indexed: 01/16/2023]
|
32
|
Adipose Tissue Hypoxia in Obesity and Its Impact on Preadipocytes and Macrophages: Hypoxia Hypothesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:305-326. [PMID: 28585205 DOI: 10.1007/978-3-319-48382-5_13] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obese subjects exhibit lower adipose tissue oxygen consumption in accordance with the lower adipose tissue blood flow. Thus, compared with lean subjects, obese subjects have 44% lower capillary density and 58% lower vascular endothelial growth factor (VEGF). The VEGF expression together with hypoxia-inducible transcription factor-1 (HIF-1) activity also requires phosphatidylinositol 3-kinase (PI3K)- and target of rapamycin (TOR)-mediated signaling. HIF-1alpha is an important signaling molecule for hypoxia to induce the inflammatory responses. Hypoxia affects a number of biological functions, such as angiogenesis, cell proliferation, apoptosis, inflammation and insulin resistance. Additionally, reactive oxygen radical (ROS) generation at mitochondria is responsible for propagation of the hypoxic signal. Actually mitochondrial ROS (mtROS) production, but not oxygen consumption is required for hypoxic HIF-1alpha protein stabilization. Adipocyte mitochondrial oxidative capacity is reduced in obese compared with non-obese adults. In this respect, mitochondrial dysfunction of adipocyte is associated with the overall adiposity. Furthermore, hypoxia also inhibits macrophage migration from the hypoxic adipose tissue. Alterations in oxygen availability of adipose tissue directly affect the macrophage polarization and are responsible from dysregulated adipocytokines production in obesity. Hypoxia also inhibits adipocyte differentiation from preadipocytes. In addition to stressed adipocytes, hypoxia contributes to immune cell immigration and activation which further aggravates adipose tissue fibrosis. Fibrosis is initiated in response to adipocyte hypertrophy in obesity.
Collapse
|
33
|
Vieira WA, Sadie-Van Gijsen H, Ferris WF. Free fatty acid G-protein coupled receptor signaling in M1 skewed white adipose tissue macrophages. Cell Mol Life Sci 2016; 73:3665-76. [PMID: 27173059 PMCID: PMC11108433 DOI: 10.1007/s00018-016-2263-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 12/13/2022]
Abstract
Obesity is associated with the establishment and maintenance of a low grade, chronically inflamed state in the white adipose tissue (WAT) of the body. The WAT macrophage population is a major cellular participant in this inflammatory process that significantly contributes to the pathophysiology of the disease, with the adipose depots of obese individuals, relative to lean counterparts, having an elevated number of macrophages that are skewed towards a pro-inflammatory phenotype. Alterations in the WAT lipid micro-environment, and specifically the availability of free fatty acids, are believed to contribute towards the obesity-related quantitative and functional changes observed in these cells. This review specifically addresses the involvement of the five G-protein coupled free fatty acid receptors which bind exogenous FFAs and signal in macrophages. Particular focus is placed on the involvement of these receptors in macrophage migration and cytokine production, two important aspects that modulate inflammation.
Collapse
Affiliation(s)
- Warren Antonio Vieira
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
| | - Hanél Sadie-Van Gijsen
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
| | - William Frank Ferris
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa.
| |
Collapse
|
34
|
Caligiuri A, Gentilini A, Marra F. Molecular Pathogenesis of NASH. Int J Mol Sci 2016; 17:ijms17091575. [PMID: 27657051 PMCID: PMC5037841 DOI: 10.3390/ijms17091575] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the main cause of chronic liver disease in the Western world and a major health problem, owing to its close association with obesity, diabetes, and the metabolic syndrome. NASH progression results from numerous events originating within the liver, as well as from signals derived from the adipose tissue and the gastrointestinal tract. In a fraction of NASH patients, disease may progress, eventually leading to advanced fibrosis, cirrhosis and hepatocellular carcinoma. Understanding the mechanisms leading to NASH and its evolution to cirrhosis is critical to identifying effective approaches for the treatment of this condition. In this review, we focus on some of the most recent data reported on the pathogenesis of NASH and its fibrogenic progression, highlighting potential targets for treatment or identification of biomarkers of disease progression.
Collapse
Affiliation(s)
- Alessandra Caligiuri
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Firenze 50121, Italy.
| | - Alessandra Gentilini
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Firenze 50121, Italy.
| | - Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Firenze 50121, Italy.
| |
Collapse
|
35
|
Gileles-Hillel A, Kheirandish-Gozal L, Gozal D. Biological plausibility linking sleep apnoea and metabolic dysfunction. Nat Rev Endocrinol 2016; 12:290-8. [PMID: 26939978 DOI: 10.1038/nrendo.2016.22] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Obstructive sleep apnoea (OSA) is a very common disorder that affects 10-25% of the general population. In the past two decades, OSA has emerged as a cardiometabolic risk factor in both paediatric and adult populations. OSA-induced metabolic perturbations include dyslipidaemia, atherogenesis, liver dysfunction and abnormal glucose metabolism. The mainstay of treatment for OSA is adenotonsillectomy in children and continuous positive airway pressure therapy in adults. Although these therapies are effective at resolving the sleep-disordered breathing component of OSA, they do not always produce beneficial effects on metabolic function. Thus, a deeper understanding of the underlying mechanisms by which OSA influences metabolic dysfunction might yield improved therapeutic approaches and outcomes. In this Review, we summarize the evidence obtained from animal models and studies of patients with OSA of potential mechanistic pathways linking the hallmarks of OSA (intermittent hypoxia and sleep fragmentation) with metabolic dysfunction. Special emphasis is given to adipose tissue dysfunction induced by sleep apnoea, which bears a striking resemblance to adipose dysfunction resulting from obesity. In addition, important gaps in current knowledge and promising lines of future investigation are identified.
Collapse
Affiliation(s)
- Alex Gileles-Hillel
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Knapp Center for Biomedical Discovery, Room 4100, 900 East 57th Street, Mailbox 4, Chicago, Illinois 60637-1470, USA
| | - Leila Kheirandish-Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Knapp Center for Biomedical Discovery, Room 4100, 900 East 57th Street, Mailbox 4, Chicago, Illinois 60637-1470, USA
| | - David Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Knapp Center for Biomedical Discovery, Room 4100, 900 East 57th Street, Mailbox 4, Chicago, Illinois 60637-1470, USA
| |
Collapse
|
36
|
Rosa TS, Simões HG, Rogero MM, Moraes MR, Denadai BS, Arida RM, Andrade MS, Silva BM. Severe Obesity Shifts Metabolic Thresholds but Does Not Attenuate Aerobic Training Adaptations in Zucker Rats. Front Physiol 2016; 7:122. [PMID: 27148063 PMCID: PMC4835489 DOI: 10.3389/fphys.2016.00122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 03/21/2016] [Indexed: 11/27/2022] Open
Abstract
Severe obesity affects metabolism with potential to influence the lactate and glycemic response to different exercise intensities in untrained and trained rats. Here we evaluated metabolic thresholds and maximal aerobic capacity in rats with severe obesity and lean counterparts at pre- and post-training. Zucker rats (obese: n = 10, lean: n = 10) were submitted to constant treadmill bouts, to determine the maximal lactate steady state, and an incremental treadmill test, to determine the lactate threshold, glycemic threshold and maximal velocity at pre and post 8 weeks of treadmill training. Velocities of the lactate threshold and glycemic threshold agreed with the maximal lactate steady state velocity on most comparisons. The maximal lactate steady state velocity occurred at higher percentage of the maximal velocity in Zucker rats at pre-training than the percentage commonly reported and used for training prescription for other rat strains (i.e., 60%) (obese = 78 ± 9% and lean = 68 ± 5%, P < 0.05 vs. 60%). The maximal lactate steady state velocity and maximal velocity were lower in the obese group at pre-training (P < 0.05 vs. lean), increased in both groups at post-training (P < 0.05 vs. pre), but were still lower in the obese group at post-training (P < 0.05 vs. lean). Training-induced increase in maximal lactate steady state, lactate threshold and glycemic threshold velocities was similar between groups (P > 0.05), whereas increase in maximal velocity was greater in the obese group (P < 0.05 vs. lean). In conclusion, lactate threshold, glycemic threshold and maximal lactate steady state occurred at similar exercise intensity in Zucker rats at pre- and post-training. Severe obesity shifted metabolic thresholds to higher exercise intensity at pre-training, but did not attenuate submaximal and maximal aerobic training adaptations.
Collapse
Affiliation(s)
- Thiago S Rosa
- Graduate Program in Translational Medicine, Federal University of São PauloSão Paulo, Brazil; Graduate Program in Physical Education and Health, Catholic University of BrasíliaBrasília, Brazil
| | - Herbert G Simões
- Graduate Program in Physical Education and Health, Catholic University of Brasília Brasília, Brazil
| | - Marcelo M Rogero
- Department of Nutrition, School of Public Health, University of São Paulo São Paulo, Brazil
| | - Milton R Moraes
- Graduate Program in Physical Education and Health, Catholic University of BrasíliaBrasília, Brazil; Department of Nephrology, Federal University of São PauloSão Paulo, Brazil
| | - Benedito S Denadai
- Human Performance Laboratory, Department of Physical Education, São Paulo State University Rio Claro, Brazil
| | - Ricardo M Arida
- Department of Physiology, Federal University of São Paulo São Paulo, Brazil
| | - Marília S Andrade
- Graduate Program in Translational Medicine, Federal University of São PauloSão Paulo, Brazil; Department of Physiology, Federal University of São PauloSão Paulo, Brazil
| | - Bruno M Silva
- Graduate Program in Translational Medicine, Federal University of São PauloSão Paulo, Brazil; Department of Physiology, Federal University of São PauloSão Paulo, Brazil
| |
Collapse
|
37
|
The Crosstalk between Hypoxia and Innate Immunity in the Development of Obesity-Related Nonalcoholic Fatty Liver Disease. BIOMED RESEARCH INTERNATIONAL 2015; 2015:319745. [PMID: 26491664 PMCID: PMC4600870 DOI: 10.1155/2015/319745] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 08/26/2015] [Accepted: 08/30/2015] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become a major health issue in western countries in parallel with the dramatic increase in the prevalence of obesity and all obesity related conditions, including respiratory diseases as obstructive sleep apnea-hypopnea syndrome (OSAHS). Interestingly, the severity of the liver damage in obesity-related NAFLD has been associated with the concomitant presence of OSAHS. In the presence of obesity, the proinflammatory state in these patients together with intermittent episodes of hypoxia, characteristic of OSAHS pathogenesis, may lead to an enhanced inflammatory response mediated by a positive feedback loop mechanism that implicates HIF-1 and NFκB. Thus, the severity of liver involvement in obese NAFLD patients with a concomitant diagnosis of OSAHS could be explained. In this review, we focus on the molecular mechanisms underlying the hepatic response to chronic intermittent hypoxia and its interaction with innate immunity in obesity-related NAFLD.
Collapse
|
38
|
Humphries F, Moynagh PN. Molecular and physiological roles of Pellino E3 ubiquitin ligases in immunity. Immunol Rev 2015; 266:93-108. [DOI: 10.1111/imr.12306] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Fiachra Humphries
- Institute of Immunology; Department of Biology; National University of Ireland Maynooth; Maynooth Ireland
| | - Paul N. Moynagh
- Institute of Immunology; Department of Biology; National University of Ireland Maynooth; Maynooth Ireland
- Centre for Infection and Immunity; School of Medicine, Dentistry and Biomedical Sciences; Queen's University Belfast; Northern Ireland UK
| |
Collapse
|
39
|
Ravaud C, Esteve D, Villageois P, Bouloumie A, Dani C, Ladoux A. IER3 Promotes Expansion of Adipose Progenitor Cells in Response to Changes in Distinct Microenvironmental Effectors. Stem Cells 2015; 33:2564-73. [PMID: 25827082 DOI: 10.1002/stem.2016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/19/2015] [Indexed: 12/23/2022]
Abstract
Adipose tissue expansion is well-orchestrated to fulfill the energy demand. It results from adipocyte hypertrophy and hyperplasia due to adipose progenitor cell (APC) expansion and differentiation. Chronic low grade inflammation and hypoxia take place in obese adipose tissue microenvironment. Both of these events were shown to impact the APC pool by promoting increased self-renewal along with a decrease in the APC differentiation potential. However, no common target has been identified so far. Here we show that the immediate early response 3 gene (IER3) is preferentially expressed in APCs and is essential for APC proliferation and self-renewal. Experiments based on RNA interference revealed that impairing IER3 expression altered cell proliferation through ERK1/2 phosphorylation and clonogenicity. IER3 expression was induced by Activin A, which plays a crucial role in adipocyte differentiation as well as by a decrease in oxygen tension through HIF1-induced transcriptional activation. Interestingly, high levels of IER3 were detected in native APCs (CD34+/CD31- cells) isolated from obese patients and conditioned media from obese adipose tissue-macrophages stimulated its expression. Overall, these results indicate that IER3 is a key player in expanding the pool of APC while highlighting the role of distinct effectors found in an obese microenvironment in this process.
Collapse
Affiliation(s)
- Christophe Ravaud
- CNRS UMR 7277, Nice, France.,University of Nice-Sophia Antipolis, Nice, France.,INSERM UMR 1091, iBV, Nice, France
| | - David Esteve
- Team 1, INSERM UMR1048, Institute of Cardiovascular and Metabolic Diseases, University of Toulouse III Paul Sabatier, Toulouse, France
| | - Phi Villageois
- CNRS UMR 7277, Nice, France.,University of Nice-Sophia Antipolis, Nice, France.,INSERM UMR 1091, iBV, Nice, France
| | - Anne Bouloumie
- Team 1, INSERM UMR1048, Institute of Cardiovascular and Metabolic Diseases, University of Toulouse III Paul Sabatier, Toulouse, France
| | - Christian Dani
- CNRS UMR 7277, Nice, France.,University of Nice-Sophia Antipolis, Nice, France.,INSERM UMR 1091, iBV, Nice, France
| | - Annie Ladoux
- CNRS UMR 7277, Nice, France.,University of Nice-Sophia Antipolis, Nice, France.,INSERM UMR 1091, iBV, Nice, France
| |
Collapse
|
40
|
Lin Q, Yun Z. The Hypoxia-Inducible Factor Pathway in Adipocytes: The Role of HIF-2 in Adipose Inflammation and Hypertrophic Cardiomyopathy. Front Endocrinol (Lausanne) 2015; 6:39. [PMID: 25852648 PMCID: PMC4369725 DOI: 10.3389/fendo.2015.00039] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/07/2015] [Indexed: 12/13/2022] Open
Abstract
Under obese conditions, adipose tissue can become oxygen-deficient or hypoxic. Extensive work has been done using various diet-induced obesity models to demonstrate an important role of hypoxia-induced signaling in adipose tissue and its impact on adipose functions related to adipogenesis, insulin sensitivity, and inflammation. We have recently identified a new mechanism connecting activation of the hypoxia-sensing pathway manifested by hypoxia-inducible factor (HIF) 2α to adipose tissue inflammation and hypertrophic cardiomyopathy. Interestingly, this observation is consistent with the clinical evidence showing that obesity is often associated with ventricular hypertrophy and dysfunction as well as congestive heart failure independent of other well-established risk factors including diabetes, hypertension, and coronary artery disease. This brief review will discuss the currently published genetic mouse models to determine the role of the HIF pathway in adipose tissue-associated diseases with a focus on the newly identified role of adipocyte HIF-2 in the development of hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Qun Lin
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Zhong Yun
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
- *Correspondence: Zhong Yun, Department of Therapeutic Radiology, Yale School of Medicine, P. O. Box 208040, New Haven, CT 06520-8040, USA e-mail:
| |
Collapse
|
41
|
Hill BG. Insights into an adipocyte whitening program. Adipocyte 2015; 4:75-80. [PMID: 26167407 DOI: 10.4161/21623945.2014.960351] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 08/26/2014] [Accepted: 08/27/2014] [Indexed: 11/19/2022] Open
Abstract
White adipose tissue plays a critical role in regulating systemic metabolism and can remodel rapidly in response to changes in nutrient availability. Nevertheless, little is known regarding the metabolic changes occurring in adipocytes during obesity. Our laboratory recently addressed this issue in a commonly used, high-fat-diet mouse model of obesity. We found remarkable changes in adipocyte metabolism that occur prior to infiltration of macrophages in expanding adipose tissue. Results of metabolomic analyses, adipose tissue respirometry, electron microscopy, and expression analyses of key genes and proteins revealed dysregulation of several metabolic pathways, loss of mitochondrial biogenetic capacity, and apparent activation of mitochondrial autophagy which were followed in time by downregulation of numerous mitochondrial proteins important for maintaining oxidative capacity. These findings demonstrate the presence of an adipocyte whitening program that may be critical for regulating adipose tissue remodeling under conditions of chronic nutrient excess.
Collapse
|
42
|
The E3 Ubiquitin Ligase Pellino3 Protects against Obesity-Induced Inflammation and Insulin Resistance. Immunity 2014; 41:973-87. [PMID: 25526310 DOI: 10.1016/j.immuni.2014.11.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 10/09/2014] [Indexed: 12/16/2022]
|
43
|
Kim M, Neinast MD, Frank AP, Sun K, Park J, Zehr JA, Vishvanath L, Morselli E, Amelotte M, Palmer BF, Gupta RK, Scherer PE, Clegg DJ. ERα upregulates Phd3 to ameliorate HIF-1 induced fibrosis and inflammation in adipose tissue. Mol Metab 2014; 3:642-51. [PMID: 25161887 PMCID: PMC4142394 DOI: 10.1016/j.molmet.2014.05.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 05/23/2014] [Accepted: 05/28/2014] [Indexed: 01/10/2023] Open
Abstract
Hypoxia Inducible Factor 1 (HIF-1) promotes fibrosis and inflammation in adipose tissues, while estrogens and Estrogen Receptor α (ERα) have the opposite effect. Here we identify an Estrogen Response Element (ERE) in the promoter of Phd3, which is a negative regulatory enzyme of HIF-1, and we demonstrate HIF-1α is ubiquitinated following 17-β estradiol (E2)/ERα mediated Phd3 transcription. Manipulating ERα in vivo increases Phd3 transcription and reduces HIF-1 activity, while addition of PHD3 ameliorates adipose tissue fibrosis and inflammation. Our findings outline a novel regulatory relationship between E2/ERα, PHD3 and HIF-1 in adipose tissues, providing a mechanistic explanation for the protective effect of E2/ERα in adipose tissue.
Collapse
Affiliation(s)
- Min Kim
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Michael D Neinast
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Aaron P Frank
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Kai Sun
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Jiyoung Park
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA ; Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology, 50 UNIST Street, Ulsan 689-798, South Korea
| | - Jordan A Zehr
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Lavanya Vishvanath
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Eugenia Morselli
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Mason Amelotte
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Biff F Palmer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Deborah J Clegg
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|