1
|
Russo L, Babboni S, Andreassi MG, Daher J, Canale P, Del Turco S, Basta G. Treating Metabolic Dysregulation and Senescence by Caloric Restriction: Killing Two Birds with One Stone? Antioxidants (Basel) 2025; 14:99. [PMID: 39857433 PMCID: PMC11763027 DOI: 10.3390/antiox14010099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Cellular senescence is a state of permanent cell cycle arrest accompanied by metabolic activity and characteristic phenotypic changes. This process is crucial for developing age-related diseases, where excessive calorie intake accelerates metabolic dysfunction and aging. Overnutrition disturbs key metabolic pathways, including insulin/insulin-like growth factor signaling (IIS), the mammalian target of rapamycin (mTOR), and AMP-activated protein kinase. The dysregulation of these pathways contributes to insulin resistance, impaired autophagy, exacerbated oxidative stress, and mitochondrial dysfunction, further enhancing cellular senescence and systemic metabolic derangements. On the other hand, dysfunctional endothelial cells and adipocytes contribute to systemic inflammation, reduced nitric oxide production, and altered lipid metabolism. Numerous factors, including extracellular vesicles, mediate pathological communication between the vascular system and adipose tissue, amplifying metabolic imbalances. Meanwhile, caloric restriction (CR) emerges as a potent intervention to counteract overnutrition effects, improve mitochondrial function, reduce oxidative stress, and restore metabolic balance. CR modulates pathways such as IIS, mTOR, and sirtuins, enhancing glucose and lipid metabolism, reducing inflammation, and promoting autophagy. CR can extend the health span and mitigate age-related diseases by delaying cellular senescence and improving healthy endothelial-adipocyte interactions. This review highlights the crosstalk between endothelial cells and adipocytes, emphasizing CR potential in counteracting overnutrition-induced senescence and restoring vascular homeostasis.
Collapse
Affiliation(s)
- Lara Russo
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Serena Babboni
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Maria Grazia Andreassi
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Jalil Daher
- Department of Biology, Faculty of Arts and Sciences, University of Balamand, El-Koura 100, Lebanon;
| | - Paola Canale
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Serena Del Turco
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| | - Giuseppina Basta
- Institute of Clinical Physiology, National Research Council, Via Moruzzi 1, 56124 Pisa, Italy; (L.R.); (S.B.); (M.G.A.); (P.C.); (G.B.)
| |
Collapse
|
2
|
Luk C, Bridge KI, Warmke N, Simmons KJ, Drozd M, Moran A, MacCannell ADV, Cheng CW, Straw S, Scragg JL, Smith J, Ozber CH, Wilkinson CG, Skromna A, Makava N, Prag HA, Simon Futers T, Brown OI, Bruns AF, Walker AM, Watt NT, Mughal R, Griffin KJ, Yuldasheva NY, Limumpornpetch S, Viswambharan H, Sukumar P, Beech DJ, Vidal-Puig A, Witte KK, Murphy MP, Hartley RC, Wheatcroft SB, Cubbon RM, Roberts LD, Kearney MT, Haywood NJ. Paracrine role of endothelial IGF-1 receptor in depot-specific adipose tissue adaptation in male mice. Nat Commun 2025; 16:170. [PMID: 39747815 PMCID: PMC11696296 DOI: 10.1038/s41467-024-54669-1] [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: 11/23/2021] [Accepted: 11/18/2024] [Indexed: 01/04/2025] Open
Abstract
During recent decades, changes in lifestyle have led to widespread nutritional obesity and its related complications. Remodelling adipose tissue as a therapeutic goal for obesity and its complications has attracted much attention and continues to be actively explored. The endothelium lines all blood vessels and is close to all cells, including adipocytes. The endothelium has been suggested to act as a paracrine organ. We explore the role of endothelial insulin-like growth factor-1 receptor (IGF-1R), as a paracrine modulator of white adipose phenotype. We show that a reduction in endothelial IGF-1R expression in the presence of high-fat feeding in male mice leads to depot-specific beneficial white adipose tissue remodelling, increases whole-body energy expenditure and enhances insulin sensitivity via a non-cell-autonomous paracrine mechanism. We demonstrate that increased endothelial malonate may be contributory and that malonate prodrugs have potentially therapeutically relevant properties in the treatment of obesity-related metabolic disease.
Collapse
Affiliation(s)
- Cheukyau Luk
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Katherine I Bridge
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Nele Warmke
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Katie J Simmons
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- School of Biomedical Sciences, Faculty of Biological Sciences & Astbury Centre, University of Leeds, Leeds, UK
| | - Michael Drozd
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Amy Moran
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Amanda D V MacCannell
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Chew W Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Sam Straw
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Jason L Scragg
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Jessica Smith
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Claire H Ozber
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Division of Gastroenterology & Surgery, Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Chloe G Wilkinson
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- North West Genomic Laboratory Hub, Manchester University NHS Foundation Trust, St Mary's Hospital, Oxford Road, Manchester, UK
| | - Anna Skromna
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Natallia Makava
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Hiran A Prag
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - T Simon Futers
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Oliver I Brown
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Alexander-Francisco Bruns
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Andrew Mn Walker
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Nicole T Watt
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Romana Mughal
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Department of Optometry and Vision Sciences, University of Huddersfield, Huddersfield, UK
| | - Kathryn J Griffin
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Nadira Y Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Sunti Limumpornpetch
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Division of Internal Medicine, Cardiology Unit, Faculty of Medicine Prince of Songkla University, Songkhla, Thailand
| | - Hema Viswambharan
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Piruthivi Sukumar
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | | | - Klaus K Witte
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK.
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| |
Collapse
|
3
|
You Z, Hu Z, Hou C, Ma C, Xu X, Zheng Y, Sun X, Ke Y, Liang J, Xie Z, Shu L, Liu Y. FABP4 facilitates epithelial-mesenchymal transition via elevating CD36 expression in glioma cells. Neoplasia 2024; 57:101050. [PMID: 39243502 PMCID: PMC11406018 DOI: 10.1016/j.neo.2024.101050] [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: 05/26/2024] [Revised: 08/17/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive brain tumor with poor prognosis. A better understanding of mechanisms concerned in glioma invasion might be critical for treatment optimization. Given that epithelial-mesenchymal transition in tumor cells is closely associated with glioma progression and recurrence, identifying pivotal mediators in GBM EMT process is urgently needed. As a member of Fatty acid binding protein (FABP) family, FABP4 serves as chaperones for free fatty acids and participates in cellular process including fatty acid uptake, transport, and metabolism. In this study, our data revealed that FABP4 expression was elevated in human GBM samples and correlated with a mesenchymal glioma subtype. Gain of function and loss of function experiments indicated that FABP4 potently rendered glioma cells increased filopodia formation and cell invasiveness. Differential expression genes analysis and GSEA in TCGA dataset revealed an EMT-related molecular signature in FABP4-mediated signaling pathways. Cell interaction analysis suggested CD36 as a potential target regulated by FABP4. Furthermore, in vitro mechanistic experiments demonstrated that FABP4-induced CD36 expression promoted EMT via non-canonical TGFβ pathways. An intracranial glioma model was constructed to assess the effect of FABP4 on tumor progression in vivo. Together, our findings demonstrated a critical role for FABP4 in the regulation invasion and EMT in GBM, and suggest that pharmacological inhibition of FABP4 may represent a promising therapeutic strategy for treatment of GBM.
Collapse
Affiliation(s)
- Zhongsheng You
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Zihao Hu
- School of Medicine, Nankai University, Tianjin, PR China
| | - Chongxian Hou
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Chengcheng Ma
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China; Department of Hematological Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, PR China; School of Medicine, Nankai University, Tianjin, PR China
| | - Xiangdong Xu
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Yaofeng Zheng
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Xinlin Sun
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Yiquan Ke
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Jianli Liang
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Zijing Xie
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Lingling Shu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China; Department of Hematological Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, PR China.
| | - Yang Liu
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China.
| |
Collapse
|
4
|
Chen S, Pan X, Gao P, Wu F. Single-nucleus transcriptome analysis identifies a novel FKBP5+ endothelial cell subtype involved in endothelial-to-mesenchymal transition in adipose tissue during aging. Biochem Biophys Res Commun 2024; 722:150157. [PMID: 38805789 DOI: 10.1016/j.bbrc.2024.150157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/03/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024]
Abstract
Age-associated adipose tissue (AT) dysfunction is multifactorial and often leads to detrimental health consequences. AT is highly vascularized and endothelial cells (ECs) has been recently identified as a key regulator in the homeostasis of AT. However, the alteration of cell composition in AT during aging and the communication between endothelial cells and adipocytes remain poorly understood. In this study, we take advantage of single nucleus RNA sequencing analysis, and discovered a group of FKBP5+ ECs specifically resident in aged AT. Of interest, FKBP5+ ECs exhibited the potential for endothelial-to-mesenchymal transition (EndoMT) and exhibited a critical role in regulating adipocytes. Furthermore, lineage tracing experiments demonstrated that ECs in aged AT tend to express FKBP5 and undergo EndoMT with progressive loss of endothelial marker. This study may provide a basis for a new mechanism of microvascular ECs-induced AT dysfunction during aging.
Collapse
Affiliation(s)
- Siyuan Chen
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxi Pan
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Pingjin Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Wu
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Clinical Research Center for Aging and Medicine, Shanghai, China.
| |
Collapse
|
5
|
Zhang YA, Li FW, Dong YX, Xie WJ, Wang HB. PPAR-γ regulates the polarization of M2 macrophages to improve the microenvironment for autologous fat grafting. FASEB J 2024; 38:e23613. [PMID: 38661048 DOI: 10.1096/fj.202400126r] [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: 01/17/2024] [Revised: 03/19/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
The unpredictable survival rate of autologous fat grafting (AFG) seriously affects its clinical application. Improving the survival rate of AFG has become an unresolved issue in plastic surgery. Peroxisome proliferator-activated receptor-γ (PPAR-γ) regulates the adipogenic differentiation of adipocytes, but the functional mechanism in AFG remains unclear. In this study, we established an animal model of AFG and demonstrated the superior therapeutic effect of PPAR-γ regulation in the process of AFG. From day 3 after fat grafting, the PPAR-γ agonist rosiglitazone group consistently showed better adipose integrity, fewer oil cysts, and fibrosis. Massive macrophage infiltration was observed after 7 days. At the same time, M2 macrophages begin to appear. At day 14, M2 macrophages gradually became the dominant cell population, which suppressed inflammation and promoted revascularization and fat regeneration. In addition, transcriptome sequencing showed that the differentially expressed genes in the Rosiglitazone group were associated with the pathways of adipose regeneration, differentiation, and angiogenesis; these results provide new ideas for clinical treatment.
Collapse
Affiliation(s)
- Ya-An Zhang
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Fang-Wei Li
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yun-Xian Dong
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Wen-Jie Xie
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Hai-Bin Wang
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
6
|
Stricher M, Vigneron P, Delbecq F, Sarde CO, Egles C. The microalga Volvox carteri as a cell supportive building block for tissue engineering. Mater Today Bio 2024; 25:101013. [PMID: 38464496 PMCID: PMC10923841 DOI: 10.1016/j.mtbio.2024.101013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 03/12/2024] Open
Abstract
Background V. carteri f. nagariensis constitutes, in its most simplified form, a cellularized spheroid built around and stabilised by a form of primitive extracellular matrix (ECM). Methods We developed a modular approach to soft tissue engineering, by compact stacking V. carteri-based building blocks. This approach is made possible by the structure and cell adhesive properties of these building blocks, which results from the composition of their algal ECM. Results A primary biocompatibility assessment demonstrated the cytocompatibility of the algal suspension, its histogenesis-promoting properties, and that it did not induce an inflammatory response in vitro. These results allowed us to consider the use of this algal suspension for soft tissue augmentation, and to initiate an in vivo biocompatibility study. V. carteri exhibited cellular fate-directing properties, causing (i) fibroblasts to take on an alkaline phosphatase+ stem-cell-like phenotype and (ii) both human adipose-derived stem cells and mouse embryonic stem cells to differentiate into preadipocytes to adipocytes. The ability of V. carteri to support histogenesis and adipogenesis was also observed in vivo by subcutaneous tissue augmentation of athymic mice, highlighting the potential of V. carteri to support or influence tissue regeneration. Conclusions We present for the first time V. carteri as an innovative and inspiring biomaterial for tissue engineering and soft tissue regeneration. Its strategies in terms of shape, structure and composition can be central in the design of a new generation of bio-inspired heterogeneous biomaterials recapitulating more appropriately the complexity of body tissues when guiding their regeneration.
Collapse
Affiliation(s)
- Mathilde Stricher
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu, CEDEX CS 60 319, 60 203, Compiègne, France
| | - Pascale Vigneron
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu, CEDEX CS 60 319, 60 203, Compiègne, France
| | - Frederic Delbecq
- Université de Technologie de Compiègne, ESCOM, TIMR (Integrated Transformations of Renewable Matter), Centre de Recherche Royallieu, CEDEX CS 60 319, 60 203, Compiègne, France
| | - Claude-Olivier Sarde
- Université de Technologie de Compiègne, ESCOM, TIMR (Integrated Transformations of Renewable Matter), Centre de Recherche Royallieu, CEDEX CS 60 319, 60 203, Compiègne, France
| | - Christophe Egles
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu, CEDEX CS 60 319, 60 203, Compiègne, France
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, 55 Rue Saint-Germain, 27 000, Évreux, France
| |
Collapse
|
7
|
Dunaway LS, Luse MA, Nyshadham S, Bulut G, Alencar GF, Chavkin NW, Cortese-Krott M, Hirschi KK, Isakson BE. Obesogenic diet disrupts tissue-specific mitochondrial gene signatures in the artery and capillary endothelium. Physiol Genomics 2024; 56:113-127. [PMID: 37982169 PMCID: PMC11281809 DOI: 10.1152/physiolgenomics.00109.2023] [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: 09/21/2023] [Revised: 11/03/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023] Open
Abstract
Endothelial cells (ECs) adapt to the unique needs of their resident tissue and metabolic perturbations, such as obesity. We sought to understand how obesity affects EC metabolic phenotypes, specifically mitochondrial gene expression. We investigated the mesenteric and adipose endothelium because these vascular beds have distinct roles in lipid homeostasis. Initially, we performed bulk RNA sequencing on ECs from mouse adipose and mesenteric vasculatures after a normal chow (NC) diet or high-fat diet (HFD) and found higher mitochondrial gene expression in adipose ECs compared with mesenteric ECs in both NC and HFD mice. Next, we performed single-cell RNA sequencing and categorized ECs as arterial, capillary, venous, or lymphatic. We found mitochondrial genes to be enriched in adipose compared with mesentery under NC conditions in artery and capillary ECs. After HFD, these genes were decreased in adipose ECs, becoming like mesenteric ECs. Transcription factor analysis revealed that peroxisome proliferator-activated receptor-γ (PPAR-γ) had high specificity in NC adipose artery and capillary ECs. These findings were recapitulated in single-nuclei RNA-sequencing data from human visceral adipose. The sum of these findings suggests that mesenteric and adipose arterial ECs metabolize lipids differently, and the transcriptional phenotype of the vascular beds converges in obesity due to downregulation of PPAR-γ in adipose artery and capillary ECs.NEW & NOTEWORTHY Using bulk and single-cell RNA sequencing on endothelial cells from adipose and mesentery, we found that an obesogenic diet induces a reduction in adipose endothelial oxidative phosphorylation gene expression, resulting in a phenotypic convergence of mesenteric and adipose endothelial cells. Furthermore, we found evidence that PPAR-γ drives this phenotypic shift. Mining of human data sets segregated based on body mass index supported these findings. These data point to novel mechanisms by which obesity induces endothelial dysfunction.
Collapse
Affiliation(s)
- Luke S Dunaway
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Melissa A Luse
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Shruthi Nyshadham
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Gamze Bulut
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Gabriel F Alencar
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Nicholas W Chavkin
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Miriam Cortese-Krott
- Department of Cardiology, Pneumology and Angiology, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Karen K Hirschi
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, United States
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| |
Collapse
|
8
|
Chaurasiya V, Pham DD, Harju J, Juuti A, Penttilä A, Emmagouni SKG, Nguyen VD, Zhang B, Perttunen S, Keskitalo S, Zhou Y, Pietiläinen KH, Haridas PAN, Olkkonen VM. Human visceral adipose tissue microvascular endothelial cell isolation and establishment of co-culture with white adipocytes to analyze cell-cell communication. Exp Cell Res 2023; 433:113819. [PMID: 37852349 DOI: 10.1016/j.yexcr.2023.113819] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/20/2023]
Abstract
Communication between adipocytes and endothelial cells (EC) is suggested to play an important role in the metabolic function of white adipose tissue. In order to generate tools to investigate in detail the physiology and communication of EC and adipocytes, a method for isolation of adipose microvascular EC from visceral adipose tissue (VAT) biopsies of subjects with obesity was developed. Moreover, mature white adipocytes were isolated from the VAT biopsies by a method adapted from a previously published Membrane aggregate adipocytes culture (MAAC) protocol. The identity and functionality of the cultivated and isolated adipose microvascular EC (AMvEC) was validated by imaging their morphology, analyses of mRNA expression, fluorescence activated cell sorting (FACS), immunostaining, low-density lipoprotein (LDL) uptake, and in vitro angiogenesis assays. Finally, we established a new trans filter co-culture system (membrane aggregate adipocyte and endothelial co-culture, MAAECC) for the analysis of communication between the two cell types. EC-adipocyte communication in this system was validated by omics analyses, revealing several altered proteins belonging to pathways such as metabolism, intracellular transport and signal transduction in adipocytes co-cultured with AMvEC. In reverse experiments, induction of several pathways including endothelial development and functions was found in AMvEC co-cultured with adipocytes. In conclusion, we developed a robust method to isolate EC from small quantities of human VAT. Furthermore, the MAAECC system established during the study enables one to study the communication between primary white adipocytes and EC or vice-versa and could also be employed for drug screening.
Collapse
Affiliation(s)
- Vaishali Chaurasiya
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland; Doctoral Programme in Biomedicine, University of Helsinki, Finland.
| | - Dan Duc Pham
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Jukka Harju
- Department of Gastrointestinal Surgery, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anne Juuti
- Department of Gastrointestinal Surgery, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anne Penttilä
- Department of Gastrointestinal Surgery, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | - Van Dien Nguyen
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK; Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, UK
| | - Birong Zhang
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK; Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, UK
| | - Sanni Perttunen
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Salla Keskitalo
- Molecular Systems Biology Research Group & Proteomics Unit, HiLIFE Helsinki Institute of Life Science, Institute of Biotechnology, University of Helsinki, Finland
| | - You Zhou
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK; Systems Immunity Research Institute, Cardiff University, Cardiff, CF14 4XN, UK
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - P A Nidhina Haridas
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland; Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland.
| |
Collapse
|
9
|
AlZaim I, de Rooij LPMH, Sheikh BN, Börgeson E, Kalucka J. The evolving functions of the vasculature in regulating adipose tissue biology in health and obesity. Nat Rev Endocrinol 2023; 19:691-707. [PMID: 37749386 DOI: 10.1038/s41574-023-00893-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/17/2023] [Indexed: 09/27/2023]
Abstract
Adipose tissue is an endocrine organ and a crucial regulator of energy storage and systemic metabolic homeostasis. Additionally, adipose tissue is a pivotal regulator of cardiovascular health and disease, mediated in part by the endocrine and paracrine secretion of several bioactive products, such as adipokines. Adipose vasculature has an instrumental role in the modulation of adipose tissue expansion, homeostasis and metabolism. The role of the adipose vasculature has been extensively explored in the context of obesity, which is recognized as a global health problem. Obesity-induced accumulation of fat, in combination with vascular rarefaction, promotes adipocyte dysfunction and induces oxidative stress, hypoxia and inflammation. It is now recognized that obesity-associated endothelial dysfunction often precedes the development of cardiovascular diseases. Investigations have revealed heterogeneity within the vascular niche and dynamic reciprocity between vascular and adipose cells, which can become dysregulated in obesity. Here we provide a comprehensive overview of the evolving functions of the vasculature in regulating adipose tissue biology in health and obesity.
Collapse
Affiliation(s)
- Ibrahim AlZaim
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Laura P M H de Rooij
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Bilal N Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich, Leipzig, Germany
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Emma Börgeson
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
10
|
Wu F, Wu F, Zhou Q, Liu X, Fei J, Zhang D, Wang W, Tao Y, Lin Y, Lin Q, Pan X, Sun K, Xie F, Bai L. A CCL2 +DPP4 + subset of mesenchymal stem cells expedites aberrant formation of creeping fat in humans. Nat Commun 2023; 14:5830. [PMID: 37730641 PMCID: PMC10511504 DOI: 10.1038/s41467-023-41418-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 09/04/2023] [Indexed: 09/22/2023] Open
Abstract
Creeping fat is a typical feature of Crohn's disease. It refers to the expansion of mesenteric adipose tissue around inflamed and fibrotic intestines and is associated with stricture formation and intestinal obstruction. In this study, we characterize creeping fat as pro-adipogenic and pro-fibrotic. Lipidomics analysis of Crohn's disease patients (sixteen males, six females) and healthy controls (five males, ten females) reveals abnormal lipid metabolism in creeping fat. Through scRNA-seq analysis on mesenteric adipose tissue from patients (five males, one female) and healthy controls (two females), we identify a CCL2+DPP4+ subset of mesenchymal stem cells that expands in creeping fat and expedites adipogenic differentiation into dystrophic adipocytes in response to CCL20+CD14+ monocytes and IL-6, leading to the formation of creeping fat. Ex vivo experiments (tissues from five males, one female) confirm that both CCL20+CD14+ monocytes and IL-6 activate DPP4+ mesenchymal stem cells towards a pro-adipogenic phenotype. This study provides a comprehensive investigation of creeping fat formation and offers a conceptual framework for discovering therapeutic targets for treatment of Crohn's disease.
Collapse
Affiliation(s)
- Fengfei Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fangting Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Zhou
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xi Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jieying Fei
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Da Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weidong Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Tao
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yubing Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiaoqiao Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Kai Sun
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fang Xie
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Lan Bai
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
11
|
Nerstedt A, Smith U. The impact of cellular senescence in human adipose tissue. J Cell Commun Signal 2023; 17:563-573. [PMID: 37195383 PMCID: PMC10409694 DOI: 10.1007/s12079-023-00769-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/07/2023] [Indexed: 05/18/2023] Open
Abstract
In the last decades the prevalence of obesity has increased dramatically, and the worldwide epidemic of obesity and related metabolic diseases has contributed to an increased interest for the adipose tissue (AT), the primary site for storage of lipids, as a metabolically dynamic and endocrine organ. Subcutaneous AT is the depot with the largest capacity to store excess energy and when its limit for storage is reached hypertrophic obesity, local inflammation, insulin resistance and ultimately type 2 diabetes (T2D) will develop. Hypertrophic AT is also associated with a dysfunctional adipogenesis, depending on the inability to recruit and differentiate new mature adipose cells. Lately, cellular senescence (CS), an aging mechanism defined as an irreversible growth arrest that occurs in response to various cellular stressors, such as telomere shortening, DNA damage and oxidative stress, has gained a lot of attention as a regulator of metabolic tissues and aging-associated conditions. The abundance of senescent cells increases not only with aging but also in hypertrophic obesity independent of age. Senescent AT is characterized by dysfunctional cells, increased inflammation, decreased insulin sensitivity and lipid storage. AT resident cells, such as progenitor cells (APC), non-proliferating mature cells and microvascular endothelial cells are affected with an increased senescence burden. Dysfunctional APC have both an impaired adipogenic and proliferative capacity. Interestingly, human mature adipose cells from obese hyperinsulinemic individuals have been shown to re-enter the cell cycle and senesce, which indicates an increased endoreplication. CS was also found to be more pronounced in mature cells from T2D individuals, compared to matched non-diabetic individuals, with decreased insulin sensitivity and adipogenic capacity. Factors associated with cellular senescence in human adipose tissue.
Collapse
Affiliation(s)
- Annika Nerstedt
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska University Hospital, Sahlgrenska Academy, University of Gothenburg, Blå Stråket 5, SE-413 45, Gothenburg, Sweden
| | - Ulf Smith
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska University Hospital, Sahlgrenska Academy, University of Gothenburg, Blå Stråket 5, SE-413 45, Gothenburg, Sweden.
| |
Collapse
|
12
|
Festa J, AlZaim I, Kalucka J. Adipose tissue endothelial cells: insights into their heterogeneity and functional diversity. Curr Opin Genet Dev 2023; 81:102055. [PMID: 37279621 DOI: 10.1016/j.gde.2023.102055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/04/2023] [Accepted: 04/28/2023] [Indexed: 06/08/2023]
Abstract
Cardiovascular disease is the leading cause of death globally. Endothelial cells (ECs), the key units of all vascular segments, have a significant impact on the health and disease of organisms. Adipose tissue is vital to cardiovascular health, therefore, understanding adipose EC (AdEC) biology is important. Recent data have highlighted the presence of distinct AdEC subpopulations that govern adipose tissue homeostasis. In addition to their role in nutrient metabolism and transport, AdECs are involved in bidirectional cellular communication with adipocytes, among other cells. These interactions are mainly mediated by paracrine factors, including noncoding RNAs. In this review, we highlight recent results showcasing the functions of AdECs in adipose tissue biology, metabolic homeostasis, and changes occurring in obesity.
Collapse
Affiliation(s)
- Joseph Festa
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus C, Denmark. https://twitter.com/@Festa_Science
| | - Ibrahim AlZaim
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus C, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark. https://twitter.com/@IbrahimAlZaim2
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus C, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
13
|
Wang Q, Wang X. The Effects of a Low Linoleic Acid/α-Linolenic Acid Ratio on Lipid Metabolism and Endogenous Fatty Acid Distribution in Obese Mice. Int J Mol Sci 2023; 24:12117. [PMID: 37569494 PMCID: PMC10419107 DOI: 10.3390/ijms241512117] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
A reduced risk of obesity and metabolic syndrome has been observed in individuals with a low intake ratio of linoleic acid/α-linolenic acid (LA/ALA). However, the influence of a low ratio of LA/ALA intake on lipid metabolism and endogenous fatty acid distribution in obese patients remains elusive. In this investigation, 8-week-old C57BL/6J mice were randomly assigned to four groups: low-fat diet (LFD) as a control, high-fat diet (HFD), high-fat diet with a low LA/ALA ratio (HFD+H3L6), and high-fat diet with a high LA/ALA ratio (HFD+L3H6) for 16 weeks. Our results show that the HFD+H3L6 diet significantly decreased the liver index of HFD mice by 3.51%, as well as the levels of triacylglycerols (TGs) and low-density lipoprotein cholesterol (LDL-C) by 15.67% and 10.02%, respectively. Moreover, the HFD+H3L6 diet reduced the pro-inflammatory cytokines interleukin-6 (IL-6) level and aspartate aminotransferase/alanine aminotransferase (AST/ALT) ratio and elevated the level of superoxide dismutase (SOD) in the liver. The HFD+H3L6 diet also resulted in the downregulation of fatty acid synthetase (FAS) and sterol regulatory element binding proteins-1c (SREBP-1c) expression and the upregulation of peroxisome proliferator-activated receptor-α (PPAR-α) and acyl-CoA oxidase 1 (ACOX1) gene expression in the liver. The low LA/ALA ratio diet led to a notable increase in the levels of ALA and its downstream derivative docosahexaenoic acid (DHA) in the erythrocyte, liver, perienteric fat, epididymal fat, perirenal fat, spleen, brain, heart, and gastrocnemius, with a strong positive correlation. Conversely, the accumulation of LA in abdominal fat was more prominent, and a high LA/ALA ratio diet exacerbated the deposition effect of LA. In conclusion, the low LA/ALA ratio not only regulated endogenous fatty acid levels but also upregulated PPAR-α and ACOX1 and downregulated SREBP-1c and FAS gene expression levels, thus maintaining lipid homeostasis. Optimizing dietary fat intake is important in studying lipid nutrition. These research findings emphasize the significance of understanding and optimizing dietary fat intake.
Collapse
Affiliation(s)
| | - Xingguo Wang
- State Key Laboratory of Food Science and Technology, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
14
|
Spinelli R, Baboota RK, Gogg S, Beguinot F, Blüher M, Nerstedt A, Smith U. Increased cell senescence in human metabolic disorders. J Clin Invest 2023; 133:e169922. [PMID: 37317964 DOI: 10.1172/jci169922] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023] Open
Abstract
Cell senescence (CS) is at the nexus between aging and associated chronic disorders, and aging increases the burden of CS in all major metabolic tissues. However, CS is also increased in adult obesity, type 2 diabetes (T2D), and nonalcoholic fatty liver disease independent of aging. Senescent tissues are characterized by dysfunctional cells and increased inflammation, and both progenitor cells and mature, fully differentiated and nonproliferating cells are afflicted. Recent studies have shown that hyperinsulinemia and associated insulin resistance (IR) promote CS in both human adipose and liver cells. Similarly, increased CS promotes cellular IR, showing their interdependence. Furthermore, the increased adipose CS in T2D is independent of age, BMI, and degree of hyperinsulinemia, suggesting premature aging. These results suggest that senomorphic/senolytic therapy may become important for treating these common metabolic disorders.
Collapse
Affiliation(s)
- Rosa Spinelli
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Translational Medical Sciences, Federico II University of Naples, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Ritesh Kumar Baboota
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Evotec International GmbH, Göttingen, Germany
| | - Silvia Gogg
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Francesco Beguinot
- Department of Translational Medical Sciences, Federico II University of Naples, Naples, Italy
- URT Genomics of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Annika Nerstedt
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulf Smith
- Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
15
|
Shu W, Wang Y, Li C, Zhang L, Zhuoma D, Yang P, Yan G, Chen C, Ba Y, Du P, Wang X. Single-cell Expression Atlas Reveals Cell Heterogeneity in the Creeping Fat of Crohn's Disease. Inflamm Bowel Dis 2023; 29:850-865. [PMID: 36715181 DOI: 10.1093/ibd/izac266] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Creeping fat (CrF) has been recognized to play a positive role in Crohn's disease (CD) progression, yet the cellular compositions within mesenteric adipose tissue (MAT) and their potential mechanism in CrF formation are poorly understood. METHODS Analysis of 10X single-cell RNA sequencing was performed on 67 064 cells from 3 pairs of surgically resected samples of CrF and their uninvolved MAT. The results were validated in another cohort with 6 paired MAT samples by immunofluorescence. RESULTS All samples manifested excellent consistency and repeatability in our study, and 10 cell types from the transcriptome atlas, including 20 clusters, were identified. In CrF, a specific vascular endothelial cell subpopulation highly expressing lipoprotein lipase was first identified, with a significantly increased proportion. This vascular endothelial cell subpopulation manifested robust peroxisome proliferator-activated receptor γ (PPARγ) transcription activity and an upregulated PPAR signaling pathway and was involved in lipid metabolism and the antibacterial response. A novel fibroblast subpopulation (FC3) with remarkable GREM1 and RFLNB expression was identified and validated to predominantly accumulate in the CrF. The FC3 was annotated as inflammation-associated fibroblasts, which are characterized by inflammatory responses and the regulation of Smad phosphorylation related to intestinal fibrosis. The trajectory of fibroblasts revealed their pro-inflammatory and profibrotic conversion tendency during CrF formation with corresponding gene dynamics. Additionally, we unprecedently dissected the different origins and functions of 6 macrophage subclusters within the myeloid compartment. CONCLUSIONS Our results uncover the cellular heterogeneity in the MAT of CD and the role of these various cellular compositions in CrF development. This comprehensive understanding of CrF provides future directions for in-depth research on and potential targets for MAT-based treatment.
Collapse
Affiliation(s)
- Weigang Shu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yongheng Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Chuanding Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Lei Zhang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Deji Zhuoma
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Pengyu Yang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Guorong Yan
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Chunqiu Chen
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Yongbing Ba
- OE Biotech Co., Ltd., Shanghai 201114, China
| | - Peng Du
- Department of Colorectal Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200092, China
| | - Xiaolei Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
16
|
Lee HJ, Lee J, Yang MJ, Kim YC, Hong SP, Kim JM, Hwang GS, Koh GY. Endothelial cell-derived stem cell factor promotes lipid accumulation through c-Kit-mediated increase of lipogenic enzymes in brown adipocytes. Nat Commun 2023; 14:2754. [PMID: 37179330 PMCID: PMC10183046 DOI: 10.1038/s41467-023-38433-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Active thermogenesis in the brown adipose tissue (BAT) facilitating the utilization of lipids and glucose is critical for maintaining body temperature and reducing metabolic diseases, whereas inactive BAT accumulates lipids in brown adipocytes (BAs), leading to BAT whitening. Although cellular crosstalk between endothelial cells (ECs) and adipocytes is essential for the transport and utilization of fatty acid in BAs, the angiocrine roles of ECs mediating this crosstalk remain poorly understood. Using single-nucleus RNA sequencing and knock-out male mice, we demonstrate that stem cell factor (SCF) derived from ECs upregulates gene expressions and protein levels of the enzymes for de novo lipogenesis, and promotes lipid accumulation by activating c-Kit in BAs. In the early phase of lipid accumulation induced by denervation or thermoneutrality, transiently expressed c-Kit on BAs increases the protein levels of the lipogenic enzymes via PI3K and AKT signaling. EC-specific SCF deletion and BA-specific c-Kit deletion attenuate the induction of the lipogenic enzymes and suppress the enlargement of lipid droplets in BAs after denervation or thermoneutrality in male mice. These data provide insight into SCF/c-Kit signaling as a regulator that promotes lipid accumulation through the increase of lipogenic enzymes in BAT when thermogenesis is inhibited.
Collapse
Affiliation(s)
- Hyuek Jong Lee
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, 34141, Republic of Korea.
| | - Jueun Lee
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 03760, Republic of Korea
| | - Myung Jin Yang
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Young-Chan Kim
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seon Pyo Hong
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, 34141, Republic of Korea
| | - Jung Mo Kim
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, 34141, Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, 03760, Republic of Korea.
- Colleage of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Gou Young Koh
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, 34141, Republic of Korea.
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
17
|
Ye J, Gao C, Liang Y, Hou Z, Shi Y, Wang Y. Characteristic and fate determination of adipose precursors during adipose tissue remodeling. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:13. [PMID: 37138165 PMCID: PMC10156890 DOI: 10.1186/s13619-023-00157-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 12/30/2022] [Indexed: 05/05/2023]
Abstract
Adipose tissues are essential for actively regulating systemic energy balance, glucose homeostasis, immune responses, reproduction, and longevity. Adipocytes maintain dynamic metabolic needs and possess heterogeneity in energy storage and supply. Overexpansion of adipose tissue, especially the visceral type, is a high risk for diabetes and other metabolic diseases. Changes in adipocytes, hypertrophy or hyperplasia, contribute to the remodeling of obese adipose tissues, accompanied by abundant immune cell accumulation, decreased angiogenesis, and aberrant extracellular matrix deposition. The process and mechanism of adipogenesis are well known, however, adipose precursors and their fate decision are only being defined with recent information available to decipher how adipose tissues generate, maintain, and remodel. Here, we discuss the key findings that identify adipose precursors phenotypically, with special emphasis on the intrinsic and extrinsic signals in instructing and regulating the fate of adipose precursors under pathophysiological conditions. We hope that the information in this review lead to novel therapeutic strategies to combat obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Jiayin Ye
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Cheng Gao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Yong Liang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Zongliu Hou
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, 650000, Yunnan, China
| | - Yufang Shi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
- The Third Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, China.
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
| |
Collapse
|
18
|
Haywood NJ, Kearney MT. Emerging paracrine functions of the endothelium in the setting of Diabetes. CURRENT OPINION IN PHYSIOLOGY 2023. [DOI: 10.1016/j.cophys.2023.100668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
19
|
Fatty Acid Metabolism in Endothelial Cell. Genes (Basel) 2022; 13:genes13122301. [PMID: 36553568 PMCID: PMC9777652 DOI: 10.3390/genes13122301] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 11/26/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
The endothelium is a monolayer of cells lining the inner blood vessels. Endothelial cells (ECs) play indispensable roles in angiogenesis, homeostasis, and immune response under normal physiological conditions, and their dysfunction is closely associated with pathologies such as cardiovascular diseases. Abnormal EC metabolism, especially dysfunctional fatty acid (FA) metabolism, contributes to the development of many diseases including pulmonary hypertension (PH). In this review, we focus on discussing the latest advances in FA metabolism in ECs under normal and pathological conditions with an emphasis on PH. We also highlight areas of research that warrant further investigation.
Collapse
|
20
|
Abstract
With obesity and its comorbidities continuing to rise, we urgently need to improve our understanding of what mechanisms trigger the white adipose tissue to become dysfunctional in response to over-feeding. The recent invent of 3D culturing models has produced several noteworthy protocols for differentiating unilocular adipocytes in vitro, promising to revolutionize the obesity research field by providing more representative adipose tissue models for such mechanistic studies. In parallel, these 3D models provide important insights to how profoundly the microenvironment influences adipocyte differentiation and morphology. This commentary highlights some of the most recent 3D models, including human unilocular vascularized adipocyte spheroids (HUVASs), developed by our lab. We discuss recent developments in the field, provide further insights to the importance of the microvasculature for adipocyte maturation, and summarize what challenges remain to be solved before we can achieve a culture model that fully recapitulates all aspects of human white adipocyte biology in vitro. Taken together, the commentary highlights important recent advances regarding 3D adipocyte culturing and underlines the many advantages these models provide over traditional 2D cultures, with the aim of convincing more laboratories to switch to 3D models.
Collapse
Affiliation(s)
- Fabiana Baganha
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ruby Schipper
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Carolina E Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
21
|
Liao ZZ, Ran L, Qi XY, Wang YD, Wang YY, Yang J, Liu JH, Xiao XH. Adipose endothelial cells mastering adipose tissues metabolic fate. Adipocyte 2022; 11:108-119. [PMID: 35067158 PMCID: PMC8786343 DOI: 10.1080/21623945.2022.2028372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/04/2022] [Accepted: 01/07/2022] [Indexed: 11/16/2022] Open
Abstract
Dynamic communication within adipose tissue depends on highly vascularized structural characteristics to maintain systemic metabolic homoeostasis. Recently, it has been noted that adipose endothelial cells (AdECs) act as essential bridges for biological information transmission between adipose-resident cells. Hence, paracrine regulators that mediate crosstalk between AdECs and adipose stromal cells were summarized. We also highlight the importance of AdECs to maintain adipocytes metabolic homoeostasis by regulating insulin sensitivity, lipid turnover and plasticity. The differential regulation of AdECs in adipose plasticity often depends on vascular density and metabolic states. Although choosing pro-angiogenic or anti-angiogenic therapies for obesity is still a matter of debate in clinical settings, the growing numbers of drugs have been confirmed to play an anti-obesity effect by affecting vascularization. Pharmacologic angiogenesis intervention has great potential as therapeutic strategies for obesity.
Collapse
Affiliation(s)
- Zhe-Zhen Liao
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li Ran
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiao-Yan Qi
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ya-Di Wang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan-Yuan Wang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jing Yang
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiang-Hua Liu
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin-Hua Xiao
- The First Affiliated Hospital of University of South China, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
22
|
Phthalate monoesters act through peroxisome proliferator-activated receptors in the mouse ovary. Reprod Toxicol 2022; 110:113-123. [PMID: 35421560 PMCID: PMC9749796 DOI: 10.1016/j.reprotox.2022.04.002] [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: 12/23/2021] [Revised: 03/21/2022] [Accepted: 04/07/2022] [Indexed: 12/16/2022]
Abstract
Widespread use of phthalates as solvents and plasticizers leads to everyday human exposure. The mechanisms by which phthalate metabolites act as ovarian toxicants are not fully understood. Thus, this study tested the hypothesis that the phthalate metabolites monononyl phthalate (MNP), monoisononyl phthalate (MiNP), mono(2-ethylhexyl) phthalate (MEHP), monobenzyl phthalate (MBzP), monobutyl phthalate (MBP), monoisobutyl phthalate (MiBP), and monoethyl phthalate (MEP) act through peroxisome proliferator-activated receptors (PPARs) in mouse granulosa cells. Primary granulosa cells were isolated from CD-1 mice and cultured with vehicle control (dimethyl sulfoxide) or MNP, MiNP, MEHP, MBzP, MBP, MiBP, or MEP (0.4-400 μM) for 24 h. Following culture, qPCR was performed for known PPAR targets, Fabp4 and Cd36. Treatment with the phthalate metabolites led to significant changes in Fabp4 and Cd36 expression relative to control in dose-dependent or nonmonotonic fashion. Primary granulosa cell cultures were also transfected with a DNA plasmid containing luciferase expressed under the control of a consensus PPAR response element. MNP, MiNP, MEHP, and MBzP caused dose-dependent changes in expression of luciferase, indicating the presence of functional endogenous PPAR receptors in the granulosa cells that respond to phthalate metabolites. The effects of phthalate metabolites on PPAR target genes were inhibited in most of the cultures by co-treatment with the PPAR-γ inhibitor, T0070907, or with the PPAR-α inhibitor, GW6471. Collectively, these data suggest that some phthalate metabolites may act through endogenous PPAR nuclear receptors in the ovary and that the differing structures of the phthalates result in different levels of activity.
Collapse
|
23
|
Rogal J, Roosz J, Teufel C, Cipriano M, Xu R, Eisler W, Weiss M, Schenke‐Layland K, Loskill P. Autologous Human Immunocompetent White Adipose Tissue-on-Chip. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104451. [PMID: 35466539 PMCID: PMC9218765 DOI: 10.1002/advs.202104451] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/03/2022] [Indexed: 05/07/2023]
Abstract
Obesity and associated diseases, such as diabetes, have reached epidemic proportions globally. In this era of "diabesity", white adipose tissue (WAT) has become a target of high interest for therapeutic strategies. To gain insights into mechanisms of adipose (patho-)physiology, researchers traditionally relied on animal models. Leveraging Organ-on-Chip technology, a microphysiological in vitro model of human WAT is introduced: a tailored microfluidic platform featuring vasculature-like perfusion that integrates 3D tissues comprising all major WAT-associated cellular components (mature adipocytes, organotypic endothelial barriers, stromovascular cells including adipose tissue macrophages) in an autologous manner and recapitulates pivotal WAT functions, such as energy storage and mobilization as well as endocrine and immunomodulatory activities. A precisely controllable bottom-up approach enables the generation of a multitude of replicates per donor circumventing inter-donor variability issues and paving the way for personalized medicine. Moreover, it allows to adjust the model's degree of complexity via a flexible mix-and-match approach. This WAT-on-Chip system constitutes the first human-based, autologous, and immunocompetent in vitro adipose tissue model that recapitulates almost full tissue heterogeneity and can become a powerful tool for human-relevant research in the field of metabolism and its associated diseases as well as for compound testing and personalized- and precision medicine applications.
Collapse
Affiliation(s)
- Julia Rogal
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGBNobelstr. 12Stuttgart70569Germany
| | - Julia Roosz
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
| | - Claudia Teufel
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| | - Madalena Cipriano
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- 3R‐Center for In vitro Models and Alternatives to Animal TestingEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| | - Raylin Xu
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGBNobelstr. 12Stuttgart70569Germany
- Harvard Medical School (HMS)25 Shattuck StBostonMA02115USA
| | - Wiebke Eisler
- Clinic for PlasticReconstructiveHand and Burn SurgeryBG Trauma CenterEberhard Karls University TübingenSchnarrenbergstraße 95Tübingen72076Germany
| | - Martin Weiss
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- Department of Women's HealthEberhard Karls University TübingenCalwerstrasse 7Tübingen72076Germany
| | - Katja Schenke‐Layland
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- Department of Medicine/CardiologyCardiovascular Research LaboratoriesDavid Geffen School of Medicine at UCLA675 Charles E. Young Drive South, MRL 3645Los AngelesCA90095USA
- Cluster of Excellence iFIT (EXC2180) “Image‐Guided and Functionally Instructed Tumor Therapies”Eberhard Karls University TuebingenRöntgenweg 11Tuebingen72076Germany
- Department for Medical Technologies and Regenerative MedicineInstitute of Biomedical EngineeringEberhard Karls University TübingenSilcherstr. 7/1Tübingen72076Germany
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- 3R‐Center for In vitro Models and Alternatives to Animal TestingEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| |
Collapse
|
24
|
De Fano M, Bartolini D, Tortoioli C, Vermigli C, Malara M, Galli F, Murdolo G. Adipose Tissue Plasticity in Response to Pathophysiological Cues: A Connecting Link between Obesity and Its Associated Comorbidities. Int J Mol Sci 2022; 23:ijms23105511. [PMID: 35628322 PMCID: PMC9141504 DOI: 10.3390/ijms23105511] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 12/10/2022] Open
Abstract
Adipose tissue (AT) is a remarkably plastic and active organ with functional pleiotropism and high remodeling capacity. Although the expansion of fat mass, by definition, represents the hallmark of obesity, the dysregulation of the adipose organ emerges as the forefront of the link between adiposity and its associated metabolic and cardiovascular complications. The dysfunctional fat displays distinct biological signatures, which include enlarged fat cells, low-grade inflammation, impaired redox homeostasis, and cellular senescence. While these events are orchestrated in a cell-type, context-dependent and temporal manner, the failure of the adipose precursor cells to form new adipocytes appears to be the main instigator of the adipose dysregulation, which, ultimately, poses a deleterious milieu either by promoting ectopic lipid overspill in non-adipose targets (i.e., lipotoxicity) or by inducing an altered secretion of different adipose-derived hormones (i.e., adipokines and lipokines). This “adipocentric view” extends the previous “expandability hypothesis”, which implies a reduced plasticity of the adipose organ at the nexus between unhealthy fat expansion and the development of obesity-associated comorbidities. In this review, we will briefly summarize the potential mechanisms by which adaptive changes to variations of energy balance may impair adipose plasticity and promote fat organ dysfunction. We will also highlight the conundrum with the perturbation of the adipose microenvironment and the development of cardio-metabolic complications by focusing on adipose lipoxidation, inflammation and cellular senescence as a novel triad orchestrating the conspiracy to adipose dysfunction. Finally, we discuss the scientific rationale for proposing adipose organ plasticity as a target to curb/prevent adiposity-linked cardio-metabolic complications.
Collapse
Affiliation(s)
- Michelatonio De Fano
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera Santa Maria Misericordia, Ospedale di Perugia, Piazzale Gambuli, 06081 Perugia, Italy; (M.D.F.); (C.T.); (C.V.); (M.M.)
| | - Desirèe Bartolini
- Department of Pharmaceutical Sciences, Human Anatomy Laboratory, University of Perugia, 06132 Perugia, Italy; (D.B.); (F.G.)
| | - Cristina Tortoioli
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera Santa Maria Misericordia, Ospedale di Perugia, Piazzale Gambuli, 06081 Perugia, Italy; (M.D.F.); (C.T.); (C.V.); (M.M.)
| | - Cristiana Vermigli
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera Santa Maria Misericordia, Ospedale di Perugia, Piazzale Gambuli, 06081 Perugia, Italy; (M.D.F.); (C.T.); (C.V.); (M.M.)
| | - Massimo Malara
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera Santa Maria Misericordia, Ospedale di Perugia, Piazzale Gambuli, 06081 Perugia, Italy; (M.D.F.); (C.T.); (C.V.); (M.M.)
| | - Francesco Galli
- Department of Pharmaceutical Sciences, Human Anatomy Laboratory, University of Perugia, 06132 Perugia, Italy; (D.B.); (F.G.)
| | - Giuseppe Murdolo
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera Santa Maria Misericordia, Ospedale di Perugia, Piazzale Gambuli, 06081 Perugia, Italy; (M.D.F.); (C.T.); (C.V.); (M.M.)
- Correspondence: ; Tel.: +39-(0)75-578-3301; Fax: +39-75-573-0855
| |
Collapse
|
25
|
The Shades of Grey in Adipose Tissue Reprogramming. Biosci Rep 2022; 42:230844. [PMID: 35211733 PMCID: PMC8905306 DOI: 10.1042/bsr20212358] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 11/22/2022] Open
Abstract
The adipose tissue (AT) has a major role in contributing to obesity-related pathologies through regulating systemic immunometabolism. The pathogenicity of the AT is underpinned by its remarkable plasticity to be reprogrammed during obesity, in the perspectives of tissue morphology, extracellular matrix (ECM) composition, angiogenesis, immunometabolic homoeostasis and circadian rhythmicity. Dysregulation in these features escalates the pathogenesis conferred by this endometabolic organ. Intriguingly, the potential to be reprogrammed appears to be an Achilles’ heel of the obese AT that can be targeted for the management of obesity and its associated comorbidities. Here, we provide an overview of the reprogramming processes of white AT (WAT), with a focus on their dynamics and pleiotropic actions over local and systemic homoeostases, followed by a discussion of potential strategies favouring therapeutic reprogramming. The potential involvement of AT remodelling in the pathogenesis of COVID-19 is also discussed.
Collapse
|
26
|
Liu R, Pugh GH, Tevonian E, Thompson K, Lauffenburger DA, Kern PA, Nikolajczyk BS. Regulatory T Cells Control Effector T Cell Inflammation in Human Prediabetes. Diabetes 2022; 71:264-274. [PMID: 34737186 PMCID: PMC8914282 DOI: 10.2337/db21-0659] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 11/01/2021] [Indexed: 02/03/2023]
Abstract
A disparate array of plasma/serum markers provides evidence for chronic inflammation in human prediabetes, a condition that is most closely replicated by standard mouse models of obesity and metaflammation. These remain largely nonactionable and contrast with our rich understanding of inflammation in human type 2 diabetes. New data show that inflammatory profiles produced by CD4+ T cells define human prediabetes as a unique inflammatory state. Regulatory T cells (Treg) control mitochondrial function and cytokine production by CD4+ effector T cells (Teff) in prediabetes and type 2 diabetes by supporting T helper (Th)17 or Th1 cytokine production, respectively. These data suggest that Treg control of Teff metabolism regulates inflammation differentially in prediabetes compared with type 2 diabetes. Queries of genes that impact mitochondrial function or pathways leading to transcription of lipid metabolism genes identified the fatty acid importer CD36 as highly expressed in Treg but not Teff from subjects with prediabetes. Pharmacological blockade of CD36 in Treg from subjects with prediabetes decreased Teff production of the Th17 cytokines that differentiate overall prediabetes inflammation. We conclude that Treg control CD4+ T cell cytokine profiles through mechanisms determined, at least in part, by host metabolic status. Furthermore, Treg CD36 uniquely promotes Th17 cytokine production by Teff in prediabetes.
Collapse
Affiliation(s)
- Rui Liu
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY
| | - Gabriella H. Pugh
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY
| | - Erin Tevonian
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA
| | - Katherine Thompson
- Dr. Bing Zhang Department of Statistics, University of Kentucky, Lexington, KY
| | | | - Philip A. Kern
- Department of Medicine, University of Kentucky, Lexington, KY
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, KY
| | - Barbara S. Nikolajczyk
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY
- Barnstable Brown Diabetes and Obesity Research Center, University of Kentucky, Lexington, KY
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY
- Corresponding author: Barbara S. Nikolajczyk,
| |
Collapse
|
27
|
Arabi T, Shafqat A, Sabbah BN, Fawzy NA, Shah H, Abdulkader H, Razak A, Sabbah AN, Arabi Z. Obesity-related kidney disease: Beyond hypertension and insulin-resistance. Front Endocrinol (Lausanne) 2022; 13:1095211. [PMID: 36726470 PMCID: PMC9884830 DOI: 10.3389/fendo.2022.1095211] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/22/2022] [Indexed: 01/19/2023] Open
Abstract
Chronic kidney disease (CKD) causes considerable morbidity, mortality, and health expenditures worldwide. Obesity is a significant risk factor for CKD development, partially explained by the high prevalence of diabetes mellitus and hypertension in obese patients. However, adipocytes also possess potent endocrine functions, secreting a myriad of cytokines and adipokines that contribute to insulin resistance and induce a chronic low-grade inflammatory state thereby damaging the kidney. CKD development itself is associated with various metabolic alterations that exacerbate adipose tissue dysfunction and insulin resistance. This adipose-renal axis is a major focus of current research, given the rising incidence of CKD and obesity. Cellular senescence is a biologic hallmark of aging, and age is another significant risk factor for obesity and CKD. An elevated senescent cell burden in adipose tissue predicts renal dysfunction in animal models, and senotherapies may alleviate these phenotypes. In this review, we discuss the direct mechanisms by which adipose tissue contributes to CKD development, emphasizing the potential clinical importance of such pathways in augmenting the care of CKD.
Collapse
Affiliation(s)
- Tarek Arabi
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- *Correspondence: Tarek Arabi,
| | - Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | | | - Hassan Shah
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Adhil Razak
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Ziad Arabi
- Division of Nephrology, Department of Medicine, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
28
|
Louis F, Sowa Y, Kitano S, Matsusaki M. High-throughput drug screening models of mature adipose tissues which replicate the physiology of patients' Body Mass Index (BMI). Bioact Mater 2022; 7:227-241. [PMID: 34466729 PMCID: PMC8379425 DOI: 10.1016/j.bioactmat.2021.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/27/2021] [Accepted: 05/07/2021] [Indexed: 12/27/2022] Open
Abstract
Obesity is a complex and incompletely understood disease, but current drug screening strategies mostly rely on immature in vitro adipose models which cannot recapitulate it properly. To address this issue, we developed a statistically validated high-throughput screening model by seeding human mature adipocytes from patients, encapsulated in physiological collagen microfibers. These drop tissues ensured the maintenance of adipocyte viability and functionality for controlling glucose and fatty acids uptake, as well as glycerol release. As such, patients' BMI and insulin sensitivity displayed a strong inverse correlation: the healthy adipocytes were associated with the highest insulin-induced glucose uptake, while insulin resistance was confirmed in the underweight and severely obese adipocytes. Insulin sensitivity recovery was possible with two type 2 diabetes treatments, rosiglitazone and melatonin. Finally, the addition of blood vasculature to the model seemed to more accurately recapitulate the in vivo physiology, with particular respect to leptin secretion metabolism.
Collapse
Affiliation(s)
- Fiona Louis
- Osaka University, Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, 2-1 Yamadaoka, Suita Osaka, 565-0871, Japan
| | - Yoshihiro Sowa
- Kyoto Prefectural University of Medicine, Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kamigyo-ku Kajii-cho, Kawaramachi-Hirokoji, Kyoto, 602-8566, Japan
- Corresponding author. Kyoto, 602-8566, Kamigyo-ku Kajii-cho, Kawaramachi-Hirokoji, Japan.
| | - Shiro Kitano
- Osaka University, Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, 2-1 Yamadaoka, Suita Osaka, 565-0871, Japan
- TOPPAN PRINTING CO., LTD., Technical Research Institute, 4-2-3 Takanodaiminami, Sugito-machi, Saitama, 345-8508, Japan
| | - Michiya Matsusaki
- Osaka University, Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, 2-1 Yamadaoka, Suita Osaka, 565-0871, Japan
- Osaka University, Graduate School of Engineering, Department of Applied Chemistry, 2-1 Yamadaoka, Suita Osaka, 565-0871, Japan
- Corresponding author. Osaka, 565-0871, 2-1 Yamadaoka, Suita, Japan.
| |
Collapse
|
29
|
Luk C, Haywood NJ, Bridge KI, Kearney MT. Paracrine Role of the Endothelium in Metabolic Homeostasis in Health and Nutrient Excess. Front Cardiovasc Med 2022; 9:882923. [PMID: 35557517 PMCID: PMC9086712 DOI: 10.3389/fcvm.2022.882923] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/04/2022] [Indexed: 02/02/2023] Open
Abstract
The vascular endothelium traditionally viewed as a simple physical barrier between the circulation and tissue is now well-established as a key organ mediating whole organism homeostasis by release of a portfolio of anti-inflammatory and pro-inflammatory vasoactive molecules. Healthy endothelium releases anti-inflammatory signaling molecules such as nitric oxide and prostacyclin; in contrast, diseased endothelium secretes pro-inflammatory signals such as reactive oxygen species, endothelin-1 and tumor necrosis factor-alpha (TNFα). Endothelial dysfunction, which has now been identified as a hallmark of different components of the cardiometabolic syndrome including obesity, type 2 diabetes and hypertension, initiates and drives the progression of tissue damage in these disorders. Recently it has become apparent that, in addition to vasoactive molecules, the vascular endothelium has the potential to secrete a diverse range of small molecules and proteins mediating metabolic processes in adipose tissue (AT), liver, skeletal muscle and the pancreas. AT plays a pivotal role in orchestrating whole-body energy homeostasis and AT dysfunction, characterized by local and systemic inflammation, is central to the metabolic complications of obesity. Thus, understanding and targeting the crosstalk between the endothelium and AT may generate novel therapeutic opportunities for the cardiometabolic syndrome. Here, we provide an overview of the role of the endothelial secretome in controlling the function of AT. The endothelial-derived metabolic regulatory factors are grouped and discussed based on their physical properties and their downstream signaling effects. In addition, we focus on the therapeutic potential of these regulatory factors in treating cardiometabolic syndrome, and discuss areas of future study of potential translatable and clinical significance. The vascular endothelium is emerging as an important paracrine/endocrine organ that secretes regulatory factors in response to nutritional and environmental cues. Endothelial dysfunction may result in imbalanced secretion of these regulatory factors and contribute to the progression of AT and whole body metabolic dysfunction. As the vascular endothelium is the first responder to local nutritional changes and adipocyte-derived signals, future work elucidating the changes in the endothelial secretome is crucial to improve our understanding of the pathophysiology of cardiometabolic disease, and in aiding our development of new therapeutic strategies to treat and prevent cardiometabolic syndrome.
Collapse
Affiliation(s)
- Cheukyau Luk
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Katherine I Bridge
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
30
|
Li R, Tan CP, Xu Y, Liu Y. Alteration of Endogenous Fatty Acids Profile and Lipid Metabolism in Rats Caused by a High‐Colleseed Oil and a High‐Sunflower Oil Diet. EUR J LIPID SCI TECH 2021. [DOI: 10.1002/ejlt.202100100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Ruizhi Li
- State Key Laboratory of Food Science and Technology School of Food Science and Technology National Engineering Research Center for Functional Food National Engineering Laboratory for Cereal Fermentation Technology Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province Jiangnan University 1800 Lihu Road Wuxi Jiangsu 214122 P. R. China
| | - Chin Ping Tan
- Department of Food Technology Faculty of Food Science and Technology Universiti Putra Malaysia Serdang Selangor 43400 Malaysia
| | - Yong‐Jiang Xu
- State Key Laboratory of Food Science and Technology School of Food Science and Technology National Engineering Research Center for Functional Food National Engineering Laboratory for Cereal Fermentation Technology Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province Jiangnan University 1800 Lihu Road Wuxi Jiangsu 214122 P. R. China
| | - Yuanfa Liu
- State Key Laboratory of Food Science and Technology School of Food Science and Technology National Engineering Research Center for Functional Food National Engineering Laboratory for Cereal Fermentation Technology Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province Jiangnan University 1800 Lihu Road Wuxi Jiangsu 214122 P. R. China
| |
Collapse
|
31
|
Hennigs JK, Matuszcak C, Trepel M, Körbelin J. Vascular Endothelial Cells: Heterogeneity and Targeting Approaches. Cells 2021; 10:2712. [PMID: 34685692 PMCID: PMC8534745 DOI: 10.3390/cells10102712] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 01/18/2023] Open
Abstract
Forming the inner layer of the vascular system, endothelial cells (ECs) facilitate a multitude of crucial physiological processes throughout the body. Vascular ECs enable the vessel wall passage of nutrients and diffusion of oxygen from the blood into adjacent cellular structures. ECs regulate vascular tone and blood coagulation as well as adhesion and transmigration of circulating cells. The multitude of EC functions is reflected by tremendous cellular diversity. Vascular ECs can form extremely tight barriers, thereby restricting the passage of xenobiotics or immune cell invasion, whereas, in other organ systems, the endothelial layer is fenestrated (e.g., glomeruli in the kidney), or discontinuous (e.g., liver sinusoids) and less dense to allow for rapid molecular exchange. ECs not only differ between organs or vascular systems, they also change along the vascular tree and specialized subpopulations of ECs can be found within the capillaries of a single organ. Molecular tools that enable selective vascular targeting are helpful to experimentally dissect the role of distinct EC populations, to improve molecular imaging and pave the way for novel treatment options for vascular diseases. This review provides an overview of endothelial diversity and highlights the most successful methods for selective targeting of distinct EC subpopulations.
Collapse
Affiliation(s)
- Jan K. Hennigs
- ENDomics Lab, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Christiane Matuszcak
- ENDomics Lab, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Martin Trepel
- Department of Hematology and Medical Oncology, University Medical Center Augsburg, 86156 Augsburg, Germany;
| | - Jakob Körbelin
- ENDomics Lab, Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| |
Collapse
|
32
|
Jin R, Hao J, Yi Y, Sauter E, Li B. Regulation of macrophage functions by FABP-mediated inflammatory and metabolic pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158964. [PMID: 33984518 PMCID: PMC8169605 DOI: 10.1016/j.bbalip.2021.158964] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022]
Abstract
Macrophages are almost everywhere in the body, where they serve pivotal functions in maintaining tissue homeostasis, remodeling, and immunoregulation. Macrophages are traditionally thought to differentiate from bone marrow-derived hematopoietic stem cells (HSCs). Emerging studies suggest that some tissue macrophages at steady state originate from embryonic precursors in the yolk sac or fetal liver and are maintained in situ by self-renewal, but bone marrow-derived monocytes can give rise to tissue macrophages in pathogenic settings, such as inflammatory injuries and cancer. Macrophages are popularly classified as Th1 cytokine (e.g. IFNγ)-activated M1 macrophages (the classical activation) or Th2 cytokine (e.g. IL-4)-activated M2 macrophages (the alternative activation). However, given the myriad arrays of stimuli macrophages may encounter from local environment, macrophages exhibit notorious heterogeneity in their phenotypes and functions. Determining the underlying metabolic pathways engaged during macrophage activation is critical for understanding macrophage phenotypic and functional adaptivity under different disease settings. Fatty acid binding proteins (FABPs) represent a family of evolutionarily conserved proteins facilitating lipid transport, metabolism and responses inside cells. More specifically, adipose-FABP (A-FABP) and epidermal-FABP (E-FABP) are highly expressed in macrophages and play a central role in integrating metabolic and inflammatory pathways. In this review we highlight how A-FABP and E-FABP are respectively upregulated in different subsets of activated macrophages and provide a unique perspective in defining macrophage phenotypic and functional heterogeneity through FABP-regulated lipid metabolic and inflammatory pathways.
Collapse
Affiliation(s)
- Rong Jin
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA; Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jiaqing Hao
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Yanmei Yi
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA; School of Basic Medical Sciences, Guangdong Medical University, Zhanjiang, China
| | - Edward Sauter
- Division of Cancer Prevention, NIH/NCI, Bethesda, MD, USA
| | - Bing Li
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
33
|
Cellular senescence and its role in white adipose tissue. Int J Obes (Lond) 2021; 45:934-943. [PMID: 33510393 DOI: 10.1038/s41366-021-00757-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/19/2020] [Accepted: 01/12/2021] [Indexed: 01/30/2023]
Abstract
Cell senescence is defined as a state of irreversible cell cycle arrest combined with DNA damage and the induction of a senescence-associated secretory phenotype (SASP). This includes increased secretion of many inflammatory agents, proteases, miRNA's, and others. Cell senescence has been widely studied in oncogenesis and has generally been considered to be protective, due to cell cycle arrest and the inhibition of proliferation. Cell senescence is also associated with ageing and extensive experimental data support its role in generating the ageing-associated phenotype. Senescent cells can also influence proximal "healthy" cells through SASPs and, e.g., inhibit normal development of progenitor/stem cells, thereby preventing tissue replacement of dying cells and reducing organ functions. Recent evidence demonstrates that SASPs may also play important roles in several chronic diseases including diabetes and cardiovascular disease. White adipose tissue (WAT) cells are highly susceptible to becoming senescent both with ageing but also with obesity and type 2 diabetes, independently of chronological age. WAT senescence is associated with inappropriate expansion (hypertrophy) of adipocytes, insulin resistance, and dyslipidemia. Major efforts have been made to identify approaches to delete senescent cells including the use of "senolytic" compounds. The most established senolytic treatment to date is the combination of dasatinib, an antagonist of the SRC family of kinases, and the antioxidant quercetin. This combination reduces cell senescence and improves chronic disorders in experimental animal models. Although only small and short-term studies have been performed in man, no severe adverse effects have been reported. Hopefully, these or other senolytic agents may provide novel ways to prevent and treat different chronic diseases in man. Here we review the current knowledge on cellular senescence in both murine and human studies. We also discuss the pathophysiological role of this process and the potential therapeutic relevance of targeting senescence selectively in WAT.
Collapse
|
34
|
Hammel JH, Bellas E. Endothelial cell crosstalk improves browning but hinders white adipocyte maturation in 3D engineered adipose tissue. Integr Biol (Camb) 2021; 12:81-89. [PMID: 32219324 DOI: 10.1093/intbio/zyaa006] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 02/07/2020] [Accepted: 02/21/2020] [Indexed: 12/13/2022]
Abstract
Central to the development of adipose tissue (AT) engineered models is the supporting vasculature. It is a key part of AT function and long-term maintenance, but the crosstalk between adipocytes and endothelial cells is not well understood. Here, we directly co-culture the two cell types at varying ratios in a 3D Type I collagen gel. Constructs were evaluated for adipocyte maturation and function and vascular network organization. Further, these constructs were treated with forskolin, a beta-adrenergic agonist, to stimulate lipolysis and browning. Adipocytes in co-cultures were found to be less mature than an adipocyte-only control, shown by smaller lipid droplets and downregulation of key adipocyte-related genes. The most extensive vascular network formation was found in the 1:1 co-culture, supported by vascular endothelial growth factor (VEGF) upregulation. After forskolin treatment, the presence of endothelial cells was shown to upregulate PPAR coactivator 1 alpha (PGC-1α) and leptin, but not uncoupling protein 1 (UCP1), suggesting a specific crosstalk that enhances early stages of browning.
Collapse
Affiliation(s)
- Jennifer H Hammel
- Department of Bioengineering, Temple University, Philadelphia, PA, USA
| | - Evangelia Bellas
- Department of Bioengineering, Temple University, Philadelphia, PA, USA
| |
Collapse
|
35
|
Baboota RK, Blüher M, Smith U. Emerging Role of Bone Morphogenetic Protein 4 in Metabolic Disorders. Diabetes 2021; 70:303-312. [PMID: 33472940 DOI: 10.2337/db20-0884] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/06/2020] [Indexed: 11/13/2022]
Abstract
Bone morphogenetic proteins (BMPs) are a group of signaling molecules that belong to the TGF-β superfamily. Initially discovered for their ability to induce bone formation, BMPs are known to play a diverse and critical array of biological roles. We here focus on recent evidence showing that BMP4 is an important regulator of white/beige adipogenic differentiation with important consequences for thermogenesis, energy homeostasis, and development of obesity in vivo. BMP4 is highly expressed in, and released by, human adipose tissue, and serum levels are increased in obesity. Recent studies have now shown BMP4 to play an important role not only for white/beige/brown adipocyte differentiation and thermogenesis but also in regulating systemic glucose homeostasis and insulin sensitivity. It also has important suppressive effects on hepatic glucose production and lipid metabolism. Cellular BMP4 signaling/action is regulated by both ambient cell/systemic levels and several endogenous and systemic BMP antagonists. Reduced BMP4 signaling/action can contribute to the development of obesity, insulin resistance, and associated metabolic disorders. In this article, we summarize the pleiotropic functions of BMP4 in the pathophysiology of these diseases and also consider the therapeutic implications of targeting BMP4 in the prevention/treatment of obesity and its associated complications.
Collapse
Affiliation(s)
- Ritesh K Baboota
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Ulf Smith
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
36
|
Dufau J, Shen JX, Couchet M, De Castro Barbosa T, Mejhert N, Massier L, Griseti E, Mouisel E, Amri EZ, Lauschke VM, Rydén M, Langin D. In vitro and ex vivo models of adipocytes. Am J Physiol Cell Physiol 2021; 320:C822-C841. [PMID: 33439778 DOI: 10.1152/ajpcell.00519.2020] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adipocytes are specialized cells with pleiotropic roles in physiology and pathology. Several types of fat cells with distinct metabolic properties coexist in various anatomically defined fat depots in mammals. White, beige, and brown adipocytes differ in their handling of lipids and thermogenic capacity, promoting differences in size and morphology. Moreover, adipocytes release lipids and proteins with paracrine and endocrine functions. The intrinsic properties of adipocytes pose specific challenges in culture. Mature adipocytes float in suspension culture due to high triacylglycerol content and are fragile. Moreover, a fully differentiated state, notably acquirement of the unilocular lipid droplet of white adipocyte, has so far not been reached in two-dimensional culture. Cultures of mouse and human-differentiated preadipocyte cell lines and primary cells have been established to mimic white, beige, and brown adipocytes. Here, we survey various models of differentiated preadipocyte cells and primary mature adipocyte survival describing main characteristics, culture conditions, advantages, and limitations. An important development is the advent of three-dimensional culture, notably of adipose spheroids that recapitulate in vivo adipocyte function and morphology in fat depots. Challenges for the future include isolation and culture of adipose-derived stem cells from different anatomic location in animal models and humans differing in sex, age, fat mass, and pathophysiological conditions. Further understanding of fat cell physiology and dysfunction will be achieved through genetic manipulation, notably CRISPR-mediated gene editing. Capturing adipocyte heterogeneity at the single-cell level within a single fat depot will be key to understanding diversities in cardiometabolic parameters among lean and obese individuals.
Collapse
Affiliation(s)
- Jérémy Dufau
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Joanne X Shen
- Karolinska Institutet, Department of Physiology and Pharmacology, Stockholm, Sweden
| | - Morgane Couchet
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | | | - Niklas Mejhert
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | - Lucas Massier
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | - Elena Griseti
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Etienne Mouisel
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | | | - Volker M Lauschke
- Karolinska Institutet, Department of Physiology and Pharmacology, Stockholm, Sweden
| | - Mikael Rydén
- Karolinska Institutet, Department of Medicine (H7), Stockholm, Sweden
| | - Dominique Langin
- Inserm, Institute of Metabolic and Cardiovascular Diseases (I2MC), UMR1297, Toulouse, France.,Faculté de Médecine, I2MC, UMR1297, Université de Toulouse, Université Paul Sabatier, Toulouse, France.,Toulouse University Hospitals, Department of Biochemistry, Toulouse, France
| |
Collapse
|
37
|
Pyrina I, Chung KJ, Michailidou Z, Koutsilieris M, Chavakis T, Chatzigeorgiou A. Fate of Adipose Progenitor Cells in Obesity-Related Chronic Inflammation. Front Cell Dev Biol 2020; 8:644. [PMID: 32760729 PMCID: PMC7372115 DOI: 10.3389/fcell.2020.00644] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/26/2020] [Indexed: 12/16/2022] Open
Abstract
Adipose progenitor cells, or preadipocytes, constitute a small population of immature cells within the adipose tissue. They are a heterogeneous group of cells, in which different subtypes have a varying degree of commitment toward diverse cell fates, contributing to white and beige adipogenesis, fibrosis or maintenance of an immature cell phenotype with proliferation capacity. Mature adipocytes as well as cells of the immune system residing in the adipose tissue can modulate the function and differentiation potential of preadipocytes in a contact- and/or paracrine-dependent manner. In the course of obesity, the accumulation of immune cells within the adipose tissue contributes to the development of a pro-inflammatory microenvironment in the tissue. Under such circumstances, the crosstalk between preadipocytes and immune or parenchymal cells of the adipose tissue may critically regulate the differentiation of preadipocytes into white adipocytes, beige adipocytes, or myofibroblasts, thereby influencing adipose tissue expansion and adipose tissue dysfunction, including downregulation of beige adipogenesis and development of fibrosis. The present review will outline the current knowledge about factors shaping cell fate decisions of adipose progenitor cells in the context of obesity-related inflammation.
Collapse
Affiliation(s)
- Iryna Pyrina
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
| | - Zoi Michailidou
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Antonios Chatzigeorgiou
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Technische Universität Dresden, Dresden, Germany
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
38
|
Munteanu R, Onaciu A, Moldovan C, Zimta AA, Gulei D, Paradiso AV, Lazar V, Berindan-Neagoe I. Adipocyte-Based Cell Therapy in Oncology: The Role of Cancer-Associated Adipocytes and Their Reinterpretation as Delivery Platforms. Pharmaceutics 2020; 12:E402. [PMID: 32354024 PMCID: PMC7284545 DOI: 10.3390/pharmaceutics12050402] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer-associated adipocytes have functional roles in tumor development through secreted adipocyte-derived factors and exosomes and also through metabolic symbiosis, where the malignant cells take up the lactate, fatty acids and glutamine produced by the neighboring adipocytes. Recent research has demonstrated the value of adipocytes as cell-based delivery platforms for drugs (or prodrugs), nucleic acids or loaded nanoparticles for cancer therapy. This strategy takes advantage of the biocompatibility of the delivery system, its ability to locate the tumor site and also the predisposition of cancer cells to come in functional contact with the adipocytes from the tumor microenvironment for metabolic sustenance. Also, their exosomal content can be used in the context of cancer stem cell reprogramming or as a delivery vehicle for different cargos, like non-coding nucleic acids. Moreover, the process of adipocytes isolation, processing and charging is quite straightforward, with minimal economical expenses. The present review comprehensively presents the role of adipocytes in cancer (in the context of obese and non-obese individuals), the main methods for isolation and characterization and also the current therapeutic applications of these cells as delivery platforms in the oncology sector.
Collapse
Affiliation(s)
- Raluca Munteanu
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Anca Onaciu
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Cristian Moldovan
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Diana Gulei
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Angelo V. Paradiso
- Oncologia Sperimentale, Istituto Tumori G Paolo II, IRCCS, 70125 Bari, Italy
| | - Vladimir Lazar
- Worldwide Innovative Network for Personalized Cancer Therapy, 94800 Villejuif, France
| | - Ioana Berindan-Neagoe
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| |
Collapse
|
39
|
Abstract
Endothelial cells line all blood vessels in vertebrates. These cells contribute to whole-body nutrient distribution in a variety of ways, including regulation of local blood flow, regulation of trans-endothelial nutrient transport, and paracrine effects. Obesity elicits dramatic whole-body nutrient redistribution, in particular of fat. We briefly review here recent progress on understanding endothelial fat transport; the impact of obesity on the endothelium; and, conversely, how endothelial function can modulate obesity.
Collapse
Affiliation(s)
- Nora Yucel
- Perelman School of Medicine, University of Pennsylvania
| | - Zolt Arany
- Perelman School of Medicine, University of Pennsylvania
| |
Collapse
|
40
|
Rekhi U, Piche JE, Immaraj L, Febbraio M. Neointimal hyperplasia: are fatty acid transport proteins a new therapeutic target? Curr Opin Lipidol 2019; 30:377-382. [PMID: 31348024 DOI: 10.1097/mol.0000000000000627] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW High-fat diets contribute to hyperlipidemia and dysregulated metabolism underlying insulin resistant states and cardiovascular diseases. Neointimal hyperplasia is a significant resulting morbidity. Increased fatty acid (FA) levels lead to dysfunctional endothelium, defined as activated, proinflammatory and prothrombotic. The purpose of this review is to assess the recent literature on the emerging concept that uptake of FA into many tissues is regulated at the endothelial level, and this in turn contributes to endothelial dysfunction, an initiating factor in insulin resistant states, atherosclerosis and neointimal hyperplasia. RECENT FINDINGS Studies support the role of endothelial FA uptake proteins as an additional level of regulation in tissue FA uptake. These proteins include CD36, FA transport proteins, FA-binding proteins and caveolin-1. In many cases, inappropriate expression of these proteins can result in a change in FA and glucose uptake, storage and utilization. Accumulation of plasma FA is one mechanism by which alterations in expression of FA uptake proteins can lead to endothelial dysfunction; changes in tissue substrate metabolism leading to inflammation are also implicated. SUMMARY Identification of the critical players and regulators can lead to therapeutic targeting to reduce endothelial dysfunction and sequela such as insulin resistance and neointimal hyperplasia.
Collapse
Affiliation(s)
- Umar Rekhi
- Department of Dentistry, Faculty of Medicine & Dentistry, University of Alberta, 7020M Katz Group Centre for Pharmacy & Health Research, Edmonton, Alberta, Canada
| | | | | | | |
Collapse
|
41
|
Ruiz HH, Díez RL, Arivazahagan L, Ramasamy R, Schmidt AM. Metabolism, Obesity, and Diabetes Mellitus. Arterioscler Thromb Vasc Biol 2019; 39:e166-e174. [PMID: 31242034 PMCID: PMC6693645 DOI: 10.1161/atvbaha.119.312005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Obesity and diabetes remain leading causes of reduced health span and life span throughout the world. Hence, it is not surprising that these areas are at the center of highly active areas of research. The identification of novel mechanisms underlying these metabolic disorders sets the stage for uncovering new potential therapeutic strategies. In this issue of Highlights in Arteriosclerosis, Thrombosis and Vascular Biology, we review recently published papers in the journal that add to our understanding of causes and consequences of obesity and diabetes and how these disorders impact metabolic function. Collectively, these studies in cultured cells to in vivo animal models to human subjects add to the growing body of evidence that both cell-intrinsic and cell-cell communication mechanisms collaborate in metabolic disorders to cause obesity, insulin resistance and diabetes and its complications.
Collapse
Affiliation(s)
- Henry H. Ruiz
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, N.Y. 10016
| | - Raquel López Díez
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, N.Y. 10016
| | - Lakshmi Arivazahagan
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, N.Y. 10016
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, N.Y. 10016
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, N.Y. 10016
| |
Collapse
|