1
|
van der Ark-Vonk EM, Puijk MV, Pasterkamp G, van der Laan SW. The Effects of FABP4 on Cardiovascular Disease in the Aging Population. Curr Atheroscler Rep 2024; 26:163-175. [PMID: 38698167 PMCID: PMC11087245 DOI: 10.1007/s11883-024-01196-5] [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] [Accepted: 03/05/2024] [Indexed: 05/05/2024]
Abstract
PURPOSE OF REVIEW Fatty acid-binding protein 4 (FABP4) plays a role in lipid metabolism and cardiovascular health. In this paper, we cover FABP4 biology, its implications in atherosclerosis from observational studies, genetic factors affecting FABP4 serum levels, and ongoing drug development to target FABP4 and offer insights into future FABP4 research. RECENT FINDINGS FABP4 impacts cells through JAK2/STAT2 and c-kit pathways, increasing inflammatory and adhesion-related proteins. In addition, FABP4 induces angiogenesis and vascular smooth muscle cell proliferation and migration. FABP4 is established as a reliable predictive biomarker for cardiovascular disease in specific at-risk groups. Genetic studies robustly link PPARG and FABP4 variants to FABP4 serum levels. Considering the potential effects on atherosclerotic lesion development, drug discovery programs have been initiated in search for potent inhibitors of FABP4. Elevated FABP4 levels indicate an increased cardiovascular risk and is causally related to acceleration of atherosclerotic disease, However, clinical trials for FABP4 inhibition are lacking, possibly due to concerns about available compounds' side effects. Further research on FABP4 genetics and its putative causal role in cardiovascular disease is needed, particularly in aging subgroups.
Collapse
Affiliation(s)
- Ellen M van der Ark-Vonk
- Central Diagnostics Laboratory, Division Laboratory, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Mike V Puijk
- Central Diagnostics Laboratory, Division Laboratory, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, Division Laboratory, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Sander W van der Laan
- Central Diagnostics Laboratory, Division Laboratory, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
2
|
Mallick R, Basak S, Das RK, Banerjee A, Paul S, Pathak S, Duttaroy AK. Fatty Acids and their Proteins in Adipose Tissue Inflammation. Cell Biochem Biophys 2024; 82:35-51. [PMID: 37794302 PMCID: PMC10867084 DOI: 10.1007/s12013-023-01185-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/06/2023]
Abstract
Chronic low-grade adipose tissue inflammation is associated with metabolic disorders. Inflammation results from the intertwined cross-talks of pro-inflammatory and anti-inflammatory pathways in the immune response of adipose tissue. In addition, adipose FABP4 levels and lipid droplet proteins are involved in systemic and tissue inflammation. Dysregulated adipocytes help infiltrate immune cells derived from bone marrow responsible for producing cytokines and chemokines. When adipose tissue expands in excess, adipocyte exhibits increased secretion of adipokines and is implicated in metabolic disturbances due to the release of free fatty acids. This review presents an emerging concept in adipose tissue fat metabolism, fatty acid handling and binding proteins, and lipid droplet proteins and their involvement in inflammatory disorders.
Collapse
Affiliation(s)
- Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Ranjit K Das
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Antara Banerjee
- Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chennai, India
| | - Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc, San Pablo, Queretaro, 76130, Mexico
| | - Surajit Pathak
- Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chennai, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, POB 1046 Blindern, Oslo, Norway.
| |
Collapse
|
3
|
Yu HC, Jeon YG, Na AY, Han CY, Lee MR, Yang JD, Yu HC, Son JB, Kim ND, Kim JB, Lee S, Bae EJ, Park BH. p21-activated kinase 4 counteracts PKA-dependent lipolysis by phosphorylating FABP4 and HSL. Nat Metab 2024; 6:94-112. [PMID: 38216738 DOI: 10.1038/s42255-023-00957-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 11/30/2023] [Indexed: 01/14/2024]
Abstract
Adipose tissue lipolysis is mediated by cAMP-protein kinase A (PKA)-dependent intracellular signalling. Here, we show that PKA targets p21-activated kinase 4 (PAK4), leading to its protein degradation. Adipose tissue-specific overexpression of PAK4 in mice attenuates lipolysis and exacerbates diet-induced obesity. Conversely, adipose tissue-specific knockout of Pak4 or the administration of a PAK4 inhibitor in mice ameliorates diet-induced obesity and insulin resistance while enhancing lipolysis. Pak4 knockout also increases energy expenditure and adipose tissue browning activity. Mechanistically, PAK4 directly phosphorylates fatty acid-binding protein 4 (FABP4) at T126 and hormone-sensitive lipase (HSL) at S565, impairing their interaction and thereby inhibiting lipolysis. Levels of PAK4 and the phosphorylation of FABP4-T126 and HSL-S565 are enhanced in the visceral fat of individuals with obesity compared to their lean counterparts. In summary, we have uncovered an important role for FABP4 phosphorylation in regulating adipose tissue lipolysis, and PAK4 inhibition may offer a therapeutic strategy for the treatment of obesity.
Collapse
Affiliation(s)
- Hwang Chan Yu
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju, Korea
| | - Yong Geun Jeon
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Ann-Yae Na
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Chang Yeob Han
- School of Pharmacy, Jeonbuk National University, Jeonju, Korea
| | - Mi Rin Lee
- Department of Surgery, Jeonbuk National University Hospital, Jeonju, Korea
| | - Jae Do Yang
- Department of Surgery, Jeonbuk National University Hospital, Jeonju, Korea
| | - Hee Chul Yu
- Department of Surgery, Jeonbuk National University Hospital, Jeonju, Korea
| | | | | | - Jae Bum Kim
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Sangkyu Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea.
| | - Eun Ju Bae
- School of Pharmacy, Jeonbuk National University, Jeonju, Korea.
| | - Byung-Hyun Park
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju, Korea.
| |
Collapse
|
4
|
Huang Y, Hertzel AV, Fish SR, Halley CL, Bohm EK, Martinez HM, Durfee CC, Sanders MA, Harris RS, Niedernhofer LJ, Bernlohr DA. TP53/p53 Facilitates Stress-Induced Exosome and Protein Secretion by Adipocytes. Diabetes 2023; 72:1560-1573. [PMID: 37347719 PMCID: PMC10588298 DOI: 10.2337/db22-1027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 06/06/2023] [Indexed: 06/24/2023]
Abstract
Besides the secretion of fatty acids, lipolytic stimulation of adipocytes results in the secretion of triglyceride-rich extracellular vesicles and some free proteins (e.g., fatty acid binding protein 4) that, in sum, affect adipose homeostasis as well as the development of metabolic disease. At the mechanistic level, lipolytic signals activate p53 in an adipose triglyceride lipase-dependent manner, and pharmacologic inhibition of p53 attenuates adipocyte-derived extracellular vesicle (AdEV) protein and FABP4 secretion. Mass spectrometry analyses of the lipolytic secretome identified proteins involved in glucose and fatty acid metabolism, translation, chaperone activities, and redox control. Consistent with a role for p53 in adipocyte protein secretion, activation of p53 by the MDM2 antagonist nutlin potentiated AdEV particles and non-AdEV protein secretion from cultured 3T3-L1 or OP9 adipocytes while the levels of FABP4 and AdEV proteins were significantly reduced in serum from p53-/- mice compared with wild-type controls. The genotoxin doxorubicin increased AdEV protein and FABP4 secretion in a p53-dependent manner and DNA repair-depleted ERCC1-/Δ-haploinsufficient mice expressed elevated p53 in adipose depots, along with significantly increased serum FABP4. In sum, these data suggest that lipolytic signals, and cellular stressors such as DNA damage, facilitate AdEV protein and FABP4 secretion by adipocytes in a p53-dependent manner. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yimao Huang
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Ann V. Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Shayla R. Fish
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Catherine L. Halley
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Ellie K. Bohm
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Hector Martell Martinez
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN
| | - Cameron C. Durfee
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Mark A. Sanders
- University Imaging Center, University of Minnesota, Minneapolis, MN
| | - Reuben S. Harris
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Laura J. Niedernhofer
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN
| | - David A. Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN
- Institute for Diabetes, Obesity and Metabolism, University of Minnesota, Minneapolis, MN
| |
Collapse
|
5
|
Inouye KE, Prentice KJ, Lee A, Wang ZB, Dominguez-Gonzalez C, Chen MX, Riveros JK, Burak MF, Lee GY, Hotamışlıgil GS. Endothelial-derived FABP4 constitutes the majority of basal circulating hormone and regulates lipolysis-driven insulin secretion. JCI Insight 2023; 8:e164642. [PMID: 37279064 PMCID: PMC10443803 DOI: 10.1172/jci.insight.164642] [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: 08/22/2022] [Accepted: 05/31/2023] [Indexed: 06/07/2023] Open
Abstract
Fatty acid binding protein 4 (FABP4) is a lipid chaperone secreted from adipocytes upon stimulation of lipolysis. Circulating FABP4 levels strongly correlate with obesity and metabolic pathologies in experimental models and humans. While adipocytes have been presumed to be the major source of hormonal FABP4, this question has not been addressed definitively in vivo. We generated mice with Fabp4 deletion in cells known to express the gene - adipocytes (Adipo-KO), endothelial cells (Endo-KO), myeloid cells (Myeloid-KO), and the whole body (Total-KO) - to examine the contribution of these cell types to basal and stimulated plasma FABP4 levels. Unexpectedly, baseline plasma FABP4 was not significantly reduced in Adipo-KO mice, whereas Endo-KO mice showed ~87% reduction versus WT controls. In contrast, Adipo-KO mice exhibited ~62% decreased induction of FABP4 responses to lipolysis, while Endo-KO mice showed only mildly decreased induction, indicating that adipocytes are the main source of increases in FABP4 during lipolysis. We did not detect any myeloid contribution to circulating FABP4. Surprisingly, despite the nearly intact induction of FABP4, Endo-KO mice showed blunted lipolysis-induced insulin secretion, identical to Total-KO mice. We conclude that the endothelium is the major source of baseline hormonal FABP4 and is required for the insulin response to lipolysis.
Collapse
Affiliation(s)
- Karen E. Inouye
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Kacey J. Prentice
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Alexandra Lee
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Zeqiu B. Wang
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Carla Dominguez-Gonzalez
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Mu Xian Chen
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Jillian K. Riveros
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - M. Furkan Burak
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Grace Y. Lee
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
| | - Gökhan S. Hotamışlıgil
- Sabri Ülker Center for Metabolic Research, Harvard T.H. Chan School of Public Health, Department of Molecular Metabolism, Boston, Massachusetts, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
6
|
Osorio-Conles Ó, Ibarzabal A, Balibrea JM, Vidal J, Ortega E, de Hollanda A. FABP4 Expression in Subcutaneous Adipose Tissue Is Independently Associated with Circulating Triglycerides in Obesity. J Clin Med 2023; 12:jcm12031013. [PMID: 36769659 PMCID: PMC9917808 DOI: 10.3390/jcm12031013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Hypertriglyceridemia (HTG) has been associated with an increased risk of pancreatitis and cardiovascular disease. Adipose tissue plays a major role in lipid metabolism, mobilization and distribution. We have compared the histological and transcriptomic profiles of the subcutaneous (SAT) and visceral (VAT) adipose tissues from subjects with severe obesity undergoing bariatric surgery with (Ob-HTG, n = 37) and without HTG (Ob-NTG, n = 67). Mean age and BMI were 51.87 ± 11.21 years, 45.78 ± 6.96 kg/m2 and 50.03 ± 10.17 years, 44.04 ± 4.69 kg/m2, respectively. The Ob-HTG group showed higher levels of glycosylated hemoglobin, fasting plasma glucose, high-sensitivity C-reactive protein and prevalence of hypertension. The degree of fibrosis was increased by 14% in SAT from the Ob-HTG group (p = 0.028), while adipocyte size distribution was comparable. Twenty genes were found differentially expressed in SAT and VAT between study groups. Among them, only SAT expression of FABP4 resulted significantly associated with circulating triglyceride levels after adjusting for other covariates and independently explained 5% of the variance in triglyceride levels in the combined model. This relationship was not found in the cohort of lean or overweight patients with normotriglyceridemia (non-Ob, n = 21). These results emphasize the contribution of SAT to triglyceride concentrations in obesity and indicate that FABP4 may be a potential drug target for the treatment of HTG.
Collapse
Affiliation(s)
- Óscar Osorio-Conles
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Monforte de Lemos Ave. 3–5, 28029 Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló Street 149, 08036 Barcelona, Spain
- Correspondence: (Ó.O.-C.); (A.d.H.); Tel.: +34-932-275-707 (ext. 2910) (Ó.O.-C.); +34-932-279-846 (A.d.H.); Fax: +34-932-275-589 (A.d.H.)
| | - Ainitze Ibarzabal
- Gastrointestinal Surgery Department, Hospital Clínic de Barcelona, Villarroel Street 170, 08036 Barcelona, Spain
| | - José María Balibrea
- Gastrointestinal Surgery Department, Hospital Clínic de Barcelona, Villarroel Street 170, 08036 Barcelona, Spain
| | - Josep Vidal
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Monforte de Lemos Ave. 3–5, 28029 Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló Street 149, 08036 Barcelona, Spain
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, Villarroel Street 170, 08036 Barcelona, Spain
| | - Emilio Ortega
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, Villarroel Street 170, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Fisiopatologia de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Monforte de Lemos Ave. 3–5, 28029 Madrid, Spain
| | - Ana de Hollanda
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló Street 149, 08036 Barcelona, Spain
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic de Barcelona, Villarroel Street 170, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Fisiopatologia de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Monforte de Lemos Ave. 3–5, 28029 Madrid, Spain
- Correspondence: (Ó.O.-C.); (A.d.H.); Tel.: +34-932-275-707 (ext. 2910) (Ó.O.-C.); +34-932-279-846 (A.d.H.); Fax: +34-932-275-589 (A.d.H.)
| |
Collapse
|
7
|
FABP4 Controls Fat Mass Expandability (Adipocyte Size and Number) through Inhibition of CD36/SR-B2 Signalling. Int J Mol Sci 2023; 24:ijms24021032. [PMID: 36674544 PMCID: PMC9867004 DOI: 10.3390/ijms24021032] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/07/2023] Open
Abstract
Adipose tissue hypertrophy during obesity plays pleiotropic effects on health. Adipose tissue expandability depends on adipocyte size and number. In mature adipocytes, lipid accumulation as triglycerides into droplets is imbalanced by lipid uptake and lipolysis. In previous studies, we showed that adipogenesis induced by oleic acid is signed by size increase and reduction of FAT/CD36 (SR-B2) activity. The present study aims to decipher the mechanisms involved in fat mass regulation by fatty acid/FAT-CD36 signalling. Human adipose stem cells, 3T3-L1, and its 3T3-MBX subclone cell lines were used in 2D cell cultures or co-cultures to monitor in real-time experiments proliferation, differentiation, lipolysis, and/or lipid uptake and activation of FAT/CD36 signalling pathways regulated by oleic acid, during adipogenesis and/or regulation of adipocyte size. Both FABP4 uptake and its induction by fatty acid-mediated FAT/CD36-PPARG gene transcription induce accumulation of intracellular FABP4, which in turn reduces FAT/CD36, and consequently exerts a negative feedback loop on FAT/CD36 signalling in both adipocytes and their progenitors. Both adipocyte size and recruitment of new adipocytes are under the control of FABP4 stores. This study suggests that FABP4 controls fat mass homeostasis.
Collapse
|
8
|
Cruciani S, Garroni G, Pala R, Coradduzza D, Cossu ML, Ginesu GC, Capobianco G, Dessole S, Ventura C, Maioli M. Metformin and vitamin D modulate adipose-derived stem cell differentiation towards the beige phenotype. Adipocyte 2022; 11:356-365. [PMID: 35734882 PMCID: PMC9235891 DOI: 10.1080/21623945.2022.2085417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) represent an ideal stem cell population for regenerative medicine. ADSC adipogenic differentiation is controlled by the activation of a specific transcriptional program, including epigenetic factors and key adipogenic genes. Under certain conditioned media, ADSCs can differentiate into several phenotypes. We previously demonstrated that bioactive molecules could counteract lipid accumulation and regulate adipogenesis, acting on inflammation and vitamin D metabolism. In the present paper, we aimed at evaluating the effect of metformin and vitamin D in targeting ADSC differentiation towards an intermediate phenotype, as beige adipocytes. We exposed ADSCs to different conditioned media and then we evaluated the levels of expression of main markers of adipogenesis, aP2, LPL and ACOT2. We also analysed the gene and protein expression of thermogenic UCP1 protein, and the expression of PARP1 and the beige specific marker TMEM26. Our results showed a novel effect of metformin and vitamin D not only in inhibiting adipogenesis, but also in inducing a specific ‘brown-like’ phenotype. These findings pave the way for their possible application in the control of de novo lipogenesis useful for the prevention of obesity and its related metabolic disorders.
Collapse
Affiliation(s)
- Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Renzo Pala
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Maria Laura Cossu
- General Surgery Unit 2 "Clinica Chirurgica" Medical, Surgical and Experimental Sciences Department, University of Sassari, Sassari, Italy
| | - Giorgio Carlo Ginesu
- General Surgery Unit 2 "Clinica Chirurgica" Medical, Surgical and Experimental Sciences Department, University of Sassari, Sassari, Italy
| | - Giampiero Capobianco
- Department of Medical, Surgical and Experimental Sciences, Gynecologic and Obstetric Clinic, University of Sassari, Sassari, Italy
| | - Salvatore Dessole
- Department of Medical, Surgical and Experimental Sciences, Gynecologic and Obstetric Clinic, University of Sassari, Sassari, Italy
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Innovation Accelerator, Consiglio Nazionale delle Ricerche, Bologna, Italy
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.,Center for Developmental Biology and Reprogramming (CEDEBIOR), Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
9
|
Bahrami-Nejad Z, Zhang ZB, Tholen S, Sharma S, Rabiee A, Zhao ML, Kraemer FB, Teruel MN. Early enforcement of cell identity by a functional component of the terminally differentiated state. PLoS Biol 2022; 20:e3001900. [PMID: 36469503 PMCID: PMC9721491 DOI: 10.1371/journal.pbio.3001900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/02/2022] [Indexed: 12/12/2022] Open
Abstract
How progenitor cells can attain a distinct differentiated cell identity is a challenging problem given the fluctuating signaling environment in which cells exist and that critical transcription factors are often not unique to a differentiation process. Here, we test the hypothesis that a unique differentiated cell identity can result from a core component of the differentiated state doubling up as a signaling protein that also drives differentiation. Using live single-cell imaging in the adipocyte differentiation system, we show that progenitor fat cells (preadipocytes) can only commit to terminally differentiate after up-regulating FABP4, a lipid buffer that is highly enriched in mature adipocytes. Upon induction of adipogenesis in mouse preadipocyte cells, we show that after a long delay, cells first abruptly start to engage a positive feedback between CEBPA and PPARG before then engaging, after a second delay, a positive feedback between FABP4 and PPARG. These sequential positive feedbacks both need to engage in order to drive PPARG levels past the threshold for irreversible differentiation. In the last step before commitment, PPARG transcriptionally increases FABP4 expression while fatty acid-loaded FABP4 increases PPARG activity. Together, our study suggests a control principle for robust cell identity whereby a core component of the differentiated state also promotes differentiation from its own progenitor state.
Collapse
Affiliation(s)
- Zahra Bahrami-Nejad
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
| | - Zhi-Bo Zhang
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
- Department of Biochemistry and the Drukier Institute for Children’s Health, Weill Cornell Medical College of Cornell University, New York, New York, United States of America
| | - Stefan Tholen
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
| | - Sanjeev Sharma
- Department of Biochemistry and the Drukier Institute for Children’s Health, Weill Cornell Medical College of Cornell University, New York, New York, United States of America
| | - Atefeh Rabiee
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
- Department of Physiology and Pharmacology, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California, United States of America
| | - Michael L. Zhao
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
| | - Fredric B. Kraemer
- Department of Medicine/Division of Endocrinology, Stanford University, Stanford, California, United States of America
- VA Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Mary N. Teruel
- Department of Chemical and Systems Biology, Stanford University, Stanford, California, United States of America
- Department of Biochemistry and the Drukier Institute for Children’s Health, Weill Cornell Medical College of Cornell University, New York, New York, United States of America
- Department of Bioengineering, Stanford University School of Medicine, Stanford, California, United States of America
- Weill Center for Metabolic Health, Division of Endocrinology, Diabetes & Metabolism, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College of Cornell University, New York, New York, United States of America
| |
Collapse
|
10
|
Zheng J, Duan Y, Zheng C, Yu J, Li F, Guo Q, Yin Y. Long-Term Protein Restriction Modulates Lipid Metabolism in White Adipose Tissues and Alters Colonic Microbiota of Shaziling Pigs. Animals (Basel) 2022; 12:ani12212944. [PMID: 36359067 PMCID: PMC9654241 DOI: 10.3390/ani12212944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/11/2022] [Accepted: 10/21/2022] [Indexed: 12/03/2022] Open
Abstract
Obesity is a matter of concern to the public. Abundant evidence has been accumulated that nutritional intervention is a promising strategy to address this health issue. The objective of this study is to investigate alterations in the lipid metabolism in white adipose tissues and the gut microbiota of Shaziling pigs challenged by long-term protein restriction. Results showed that compared with the control group, reducing the protein level by 20% (−20%) increased the mRNA abundance of FABP4 in white adipose tissues (p < 0.05). This occurred in conjunction with increases in PPARγ protein expression. Conversely, the protein expression of C/EBPα was reduced in the −20% group (p < 0.05). Moreover, the −20% group had increased/decreased phosphorylation of AMPKα/mTOR, respectively (p < 0.05). As for the colonic gut microbiota, a 20% reduction in the protein level led to increased Lachnospiraceae XPB1014 group abundance at the genus level (p < 0.01). Collectively, these results indicated that a 20% protein reduction could modulate lipid metabolism and alter the colonic microbiota of Shaziling pigs, an approach which might be translated into a treatment for obesity.
Collapse
Affiliation(s)
- Jie Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Yehui Duan
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
- Correspondence: (Y.D.); (Y.Y.)
| | - Changbing Zheng
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Jiayi Yu
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Fengna Li
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Qiuping Guo
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
| | - Yulong Yin
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100039, China
- Correspondence: (Y.D.); (Y.Y.)
| |
Collapse
|
11
|
Watkins OC, Yong HEJ, Mah TKL, Cracknell-Hazra VKB, Pillai RA, Selvam P, Sharma N, Cazenave-Gassiot A, Bendt AK, Godfrey KM, Lewis RM, Wenk MR, Chan SY. Sex-Dependent Regulation of Placental Oleic Acid and Palmitic Acid Metabolism by Maternal Glycemia and Associations with Birthweight. Int J Mol Sci 2022; 23:ijms23158685. [PMID: 35955818 PMCID: PMC9369035 DOI: 10.3390/ijms23158685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 01/27/2023] Open
Abstract
Pregnancy complications such as maternal hyperglycemia increase perinatal mortality and morbidity, but risks are higher in males than in females. We hypothesized that fetal sex-dependent differences in placental palmitic-acid (PA) and oleic-acid (OA) metabolism influence such risks. Placental explants (n = 22) were incubated with isotope-labeled fatty acids (13C-PA or 13C-OA) for 24 or 48 h and the production of forty-seven 13C-PA lipids and thirty-seven 13C-OA lipids quantified by LCMS. Linear regression was used to investigate associations between maternal glycemia, BMI and fetal sex with 13C lipids, and between 13C lipids and birthweight centile. Placental explants from females showed greater incorporation of 13C-OA and 13C-PA into almost all lipids compared to males. Fetal sex also influenced relationships with maternal glycemia, with many 13C-OA and 13C-PA acylcarnitines, 13C-PA-diacylglycerols and 13C-PA phospholipids positively associated with glycemia in females but not in males. In contrast, several 13C-OA triacylglycerols and 13C-OA phospholipids were negatively associated with glycemia in males but not in females. Birthweight centile in females was positively associated with six 13C-PA and three 13C-OA lipids (mainly acylcarnitines) and was negatively associated with eight 13C-OA lipids, while males showed few associations. Fetal sex thus influences placental lipid metabolism and could be a key modulator of the impact of maternal metabolic health on perinatal outcomes, potentially contributing toward sex-specific adaptions in which females prioritize survival.
Collapse
Affiliation(s)
- Oliver C. Watkins
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Hannah E. J. Yong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore 117609, Singapore
| | - Tania Ken Lin Mah
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore 117609, Singapore
| | - Victoria K. B. Cracknell-Hazra
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore 117609, Singapore
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO17 1BJ, UK
| | - Reshma Appukuttan Pillai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Preben Selvam
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Neha Sharma
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Amaury Cazenave-Gassiot
- Department of Biochemistry and Precision Medicine TRP, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore 119077, Singapore
| | - Anne K. Bendt
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore 119077, Singapore
| | - Keith M. Godfrey
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO17 1BJ, UK
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO17 1BJ, UK
| | - Rohan M. Lewis
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO17 1BJ, UK
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Markus R. Wenk
- Department of Biochemistry and Precision Medicine TRP, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore 119077, Singapore
| | - Shiao-Yng Chan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore 117609, Singapore
- Correspondence:
| |
Collapse
|
12
|
Basak S, Mallick R, Banerjee A, Pathak S, Duttaroy AK. Cytoplasmic fatty acid-binding proteins in metabolic diseases and cancers. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 132:143-174. [PMID: 36088074 DOI: 10.1016/bs.apcsb.2022.05.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytoplasmic fatty acid-binding proteins (FABPs) are multipurpose proteins that can modulate lipid fluxes, trafficking, signaling, and metabolism. FABPs regulate metabolic and inflammatory pathways, its inhibition can improve type 2 diabetes mellitus and atherosclerosis. In addition, FABPs are involved in obesity, metabolic disease, cardiac dysfunction, and cancers. FABPs are promising tissue biomarkers in solid tumors for diagnostic and/or prognostic targets for novel therapeutic strategies. The signaling responsive elements of FABPs and determinants of FABP-mediated functions may be exploited in preventing or treating these diseases.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| | - Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
13
|
Adipocyte Phenotype Flexibility and Lipid Dysregulation. Cells 2022; 11:cells11050882. [PMID: 35269504 PMCID: PMC8909878 DOI: 10.3390/cells11050882] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/22/2022] [Accepted: 03/01/2022] [Indexed: 12/04/2022] Open
Abstract
The prevalence of obesity and associated cardiometabolic diseases continues to rise, despite efforts to improve global health. The adipose tissue is now regarded as an endocrine organ since its multitude of secretions, lipids chief among them, regulate systemic functions. The loss of normal adipose tissue phenotypic flexibility, especially related to lipid homeostasis, appears to trigger cardiometabolic pathogenesis. The goal of this manuscript is to review lipid balance maintenance by the lean adipose tissue’s propensity for phenotype switching, obese adipose tissue’s narrower range of phenotype flexibility, and what initial factors account for the waning lipid regulatory capacity. Metabolic, hypoxic, and inflammatory factors contribute to the adipose tissue phenotype being made rigid. A better grasp of normal adipose tissue function provides the necessary context for recognizing the extent of obese adipose tissue dysfunction and gaining insight into how pathogenesis evolves.
Collapse
|
14
|
Gruber N, Rathaus M, Ron I, Livne R, Sheinvald S, Barhod E, Hemi R, Tirosh A, Pinhas-Hamiel O, Tirosh A. Fatty acid-binding protein 4: a key regulator of ketoacidosis in new-onset type 1 diabetes. Diabetologia 2022; 65:366-374. [PMID: 34806114 DOI: 10.1007/s00125-021-05606-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/06/2021] [Indexed: 02/05/2023]
Abstract
AIMS/HYPOTHESIS Fatty acid-binding protein 4 (FABP4) is an adipokine with a key regulatory role in glucose and lipid metabolism. We prospectively evaluated the role of FABP4 in the pathophysiology of diabetic ketoacidosis (DKA) in new-onset type 1 diabetes. METHODS Clinical and laboratory data were prospectively collected from consecutive children presenting with new-onset type 1 diabetes. In addition to blood chemistry and gases, insulin, C-peptide, serum FABP4 and NEFA were collected upon presentation and 48 h after initiation of insulin treatment. In a mouse model of type 1 diabetes, glucose, insulin, β-hydroxybutyrate and weight were compared between FABP4 knockout (Fabp4-/-) and wild-type (WT) mice. RESULTS Included were 33 children (mean age 9.3 ± 3.5 years, 52% male), of whom 14 (42%) presented with DKA. FABP4 levels were higher in the DKA group compared with the non-DKA group (median [IQR] 10.1 [7.9-14.2] ng/ml vs 6.3 [3.9-7] ng/ml, respectively; p = 0.005). The FABP4 level was positively correlated with HbA1c at presentation and inversely correlated with venous blood pH and bicarbonate levels (p < 0.05 for all). Following initiation of insulin therapy, a marked reduction in FABP4 was observed in all children. An FABP4 level of 7.22 ng/ml had a sensitivity of 86% and a specificity of 78% for the diagnosis of DKA, with an area under the receiver operating characteristic curve of 0.78 (95% CI 0.6, 0.95; p = 0.008). In a streptozotocin-induced diabetes mouse model, Fabp4-/- mice exhibited marked hypoinsulinaemia and hyperglycaemia similar to WT mice but displayed no significant increase in β-hydroxybutyrate and were protected from ketoacidosis. CONCLUSIONS/INTERPRETATION FABP4 is suggested to be a necessary regulator of ketogenesis in insulin-deficient states.
Collapse
Affiliation(s)
- Noah Gruber
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Moran Rathaus
- The Dalia and David Arabov Diabetes Research Center, Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Tel-Hashomer, Israel
| | - Idit Ron
- The Dalia and David Arabov Diabetes Research Center, Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Tel-Hashomer, Israel
| | - Rinat Livne
- The Dalia and David Arabov Diabetes Research Center, Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Tel-Hashomer, Israel
| | - Sharon Sheinvald
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Ehud Barhod
- The Dalia and David Arabov Diabetes Research Center, Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Tel-Hashomer, Israel
| | - Rina Hemi
- The Dalia and David Arabov Diabetes Research Center, Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Tel-Hashomer, Israel
| | - Amit Tirosh
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- The Dalia and David Arabov Diabetes Research Center, Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Tel-Hashomer, Israel
| | - Orit Pinhas-Hamiel
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Amir Tirosh
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
- The Dalia and David Arabov Diabetes Research Center, Division of Endocrinology, Diabetes and Metabolism, Sheba Medical Center, Tel-Hashomer, Israel.
| |
Collapse
|
15
|
Unveiling the Role of the Fatty Acid Binding Protein 4 in the Metabolic-Associated Fatty Liver Disease. Biomedicines 2022; 10:biomedicines10010197. [PMID: 35052876 PMCID: PMC8773613 DOI: 10.3390/biomedicines10010197] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 02/04/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD), the main cause of chronic liver disease worldwide, is a progressive disease ranging from fatty liver to steatohepatitis (metabolic-associated steatohepatitis; MASH). Nevertheless, it remains underdiagnosed due to the lack of effective non-invasive methods for its diagnosis and staging. Although MAFLD has been found in lean individuals, it is closely associated with obesity-related conditions. Adipose tissue is the main source of liver triglycerides and adipocytes act as endocrine organs releasing a large number of adipokines and pro-inflammatory mediators involved in MAFLD progression into bloodstream. Among the adipocyte-derived molecules, fatty acid binding protein 4 (FABP4) has been recently associated with fatty liver and additional features of advanced stages of MAFLD. Additionally, emerging data from preclinical studies propose FABP4 as a causal actor involved in the disease progression, rather than a mere biomarker for the disease. Therefore, the FABP4 regulation could be considered as a potential therapeutic strategy to MAFLD. Here, we review the current knowledge of FABP4 in MAFLD, as well as its potential role as a therapeutic target for this disease.
Collapse
|
16
|
Sprenger S, Woldemariam T, Kotchoni S, Elshabrawy HA, Chaturvedi LS. Lemongrass essential oil and its major constituent citral isomers modulate adipogenic gene expression in 3T3-L1 cells. J Food Biochem 2022; 46:e14037. [PMID: 34981531 DOI: 10.1111/jfbc.14037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 10/07/2021] [Accepted: 10/27/2021] [Indexed: 12/19/2022]
Abstract
Obesity is a predisposing factor to diseases such as diabetes mellitus, hypertension, and coronary artery disease. Lemongrass essential oil (LEO), from Cymbopogon flexuosus, possesses numerous therapeutic properties including modulation of obesity in vivo. This experiment investigated the effect of LEO and its major components citral (3,7-dimethyl-2,6-octadienal), citral dimethyl acetal (1,1-dimethoxy-3,7-dimethylocta-2,6-diene), and citral diethyl acetal (1,1-diethoxy-3,7-dimethylocta-2,6-diene) in modulation of adipogenesis and genetic expression in adipocytes. Adipogenesis was induced from murine 3T3-L1 preadipocytes procured from ATCC and maintained in Dulbecco's modified Eagle's medium (DMEM) enriched with calf serum. Differentiation was conducted using DMEM enriched with 10% fetal bovine serum, Dexamethasone 0.25 µM, 3-isobutyl-methylxanthine 0.5 mM, and insulin 10 mg/ml for 2 days, followed by 5 days of insulin 10 mg/ml alone. Samples were subjected to experimental treatments at a concentration of 2.5 × 10-3 . Intracellular triglycerides were quantified and photomicrographs were obtained following Oil red O (ORO) staining procedure. Total ribonucleic acid was extracted and expression of genes effecting in lipid metabolism were quantitated using real-time polymerase chain reaction. ORO staining procedure and spectrophotometric analysis demonstrated decreased lipid accumulation following treatments. LEO and its major constituents significantly inhibited expression of sterol response binding protein 2, cluster of differentiation 36, fatty acid binding protein 4, and peripilin. These results indicate modulation of lipid accumulation through decreased lipid uptake, increased lipolysis, decreased differentiation, and downregulated lipid biosynthesis. This investigation suggests that LEO and its constituents exert effects on adipocyte metabolism and are important for understanding metabolic disease. Further investigation is required to elucidate the degree that each mechanism implicated contributes to the observed effect.
Collapse
Affiliation(s)
- Steven Sprenger
- Department of Basic Science, California Northstate University College of Medicine, Elk Grove, California, USA
| | - Tibebe Woldemariam
- Department of Pharmaceutical & Biomedical Science, California Northstate University College of Pharmacy, Elk Grove, California, USA
| | - Simeon Kotchoni
- Department of Pharmaceutical & Biomedical Science, California Northstate University College of Pharmacy, Elk Grove, California, USA
| | - Hatem A Elshabrawy
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, Texas, USA
| | - Lakshmi Shankar Chaturvedi
- Department of Pharmaceutical & Biomedical Sciences, College of Pharmacy, Basic Science and Surgery, California Northstate University College of Medicine, Elk Grove, California, USA
| |
Collapse
|
17
|
Yoon H, Shaw JL, Haigis MC, Greka A. Lipid metabolism in sickness and in health: Emerging regulators of lipotoxicity. Mol Cell 2021; 81:3708-3730. [PMID: 34547235 PMCID: PMC8620413 DOI: 10.1016/j.molcel.2021.08.027] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022]
Abstract
Lipids play crucial roles in signal transduction, contribute to the structural integrity of cellular membranes, and regulate energy metabolism. Questions remain as to which lipid species maintain metabolic homeostasis and which disrupt essential cellular functions, leading to metabolic disorders. Here, we discuss recent advances in understanding lipid metabolism with a focus on catabolism, synthesis, and signaling. Technical advances, including functional genomics, metabolomics, lipidomics, lipid-protein interaction maps, and advances in mass spectrometry, have uncovered new ways to prioritize molecular mechanisms mediating lipid function. By reviewing what is known about the distinct effects of specific lipid species in physiological pathways, we provide a framework for understanding newly identified targets regulating lipid homeostasis with implications for ameliorating metabolic diseases.
Collapse
Affiliation(s)
- Haejin Yoon
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Ludwig Center for Cancer Research at Harvard, Boston, MA 02115, USA
| | - Jillian L Shaw
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Ludwig Center for Cancer Research at Harvard, Boston, MA 02115, USA.
| | - Anna Greka
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
18
|
A-FABP in Metabolic Diseases and the Therapeutic Implications: An Update. Int J Mol Sci 2021; 22:ijms22179386. [PMID: 34502295 PMCID: PMC8456319 DOI: 10.3390/ijms22179386] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022] Open
Abstract
Adipocyte fatty acid-binding protein (A-FABP), which is also known as ap2 or FABP4, is a fatty acid chaperone that has been further defined as a fat-derived hormone. It regulates lipid homeostasis and is a key mediator of inflammation. Circulating levels of A-FABP are closely associated with metabolic syndrome and cardiometabolic diseases with imminent diagnostic and prognostic significance. Numerous animal studies have elucidated the potential underlying mechanisms involving A-FABP in these diseases. Recent studies demonstrated its physiological role in the regulation of adaptive thermogenesis and its pathological roles in ischemic stroke and liver fibrosis. Due to its implication in various diseases, A-FABP has become a promising target for the development of small molecule inhibitors and neutralizing antibodies for disease treatment. This review summarizes the clinical and animal findings of A-FABP in the pathogenesis of cardio-metabolic diseases in recent years. The underlying mechanism and its therapeutic implications are also highlighted.
Collapse
|
19
|
Yang X, Chen Q, Ouyang Q, Rong P, Feng W, Quan C, Li M, Jiang Q, Liang H, Zhao TJ, Wang HY, Chen S. Tissue-Specific Splicing and Dietary Interaction of a Mutant As160 Allele Determine Muscle Metabolic Fitness in Rodents. Diabetes 2021; 70:1826-1842. [PMID: 33980689 DOI: 10.2337/db21-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 05/06/2021] [Indexed: 11/13/2022]
Abstract
Ethnic groups are physiologically and genetically adapted to their diets. Inuit bear a frequent AS160R684X mutation that causes type 2 diabetes. Whether this mutation evolutionarily confers adaptation in Inuit and how it causes metabolic disorders upon dietary changes are unknown due to limitations in human studies. Here, we develop a genetically modified rat model bearing an orthologous AS160R693X mutation, which mimics human patients exhibiting postprandial hyperglycemia and hyperinsulinemia. Importantly, a sugar-rich diet aggravates metabolic abnormalities in AS160R693X rats. The AS160R693X mutation diminishes a dominant long-variant AS160 without affecting a minor short-variant AS160 in skeletal muscle, which suppresses muscle glucose utilization but induces fatty acid oxidation. This fuel switch suggests a possible adaptation in Inuit who traditionally had lipid-rich hypoglycemic diets. Finally, induction of the short-variant AS160 restores glucose utilization in rat myocytes and a mouse model. Our findings have implications for development of precision treatments for patients bearing the AS160R684X mutation.
Collapse
Affiliation(s)
- Xinyu Yang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Qiaoli Chen
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Qian Ouyang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Ping Rong
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Weikuan Feng
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Chao Quan
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Min Li
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive Surgery, Drum Tower Hospital, School of Medicine, Nanjing University, China
| | - Hui Liang
- Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Tong-Jin Zhao
- Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Hong Yu Wang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| | - Shuai Chen
- MOE Key Laboratory of Model Animal for Disease Study, Department of Endocrinology, Nanjing Drum Tower Hospital, and Model Animal Research Center, School of Medicine, Nanjing University, Nanjing
| |
Collapse
|
20
|
Scaglia N, Frontini-López YR, Zadra G. Prostate Cancer Progression: as a Matter of Fats. Front Oncol 2021; 11:719865. [PMID: 34386430 PMCID: PMC8353450 DOI: 10.3389/fonc.2021.719865] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
Advanced prostate cancer (PCa) represents the fifth cause of cancer death worldwide. Although survival has improved with second-generation androgen signaling and Parp inhibitors, the benefits are not long-lasting, and new therapeutic approaches are sorely needed. Lipids and their metabolism have recently reached the spotlight with accumulating evidence for their role as promoters of PCa development, progression, and metastasis. As a result, interest in targeting enzymes/transporters involved in lipid metabolism is rapidly growing. Moreover, the use of lipogenic signatures to predict prognosis and resistance to therapy has been recently explored with promising results. Despite the well-known association between obesity with PCa lethality, the underlying mechanistic role of diet/obesity-derived metabolites has only lately been unveiled. Furthermore, the role of lipids as energy source, building blocks, and signaling molecules in cancer cells has now been revisited and expanded in the context of the tumor microenvironment (TME), which is heavily influenced by the external environment and nutrient availability. Here, we describe how lipids, their enzymes, transporters, and modulators can promote PCa development and progression, and we emphasize the role of lipids in shaping TME. In a therapeutic perspective, we describe the ongoing efforts in targeting lipogenic hubs. Finally, we highlight studies supporting dietary modulation in the adjuvant setting with the purpose of achieving greater efficacy of the standard of care and of synthetic lethality. PCa progression is "a matter of fats", and the more we understand about the role of lipids as key players in this process, the better we can develop approaches to counteract their tumor promoter activity while preserving their beneficial properties.
Collapse
Affiliation(s)
- Natalia Scaglia
- Biochemistry Research Institute of La Plata "Professor Doctor Rodolfo R. Brenner" (INIBIOLP), National University of La Plata/National Council of Scientific and Technical Research of Argentina, La Plata, Argentina
| | - Yesica Romina Frontini-López
- Biochemistry Research Institute of La Plata "Professor Doctor Rodolfo R. Brenner" (INIBIOLP), National University of La Plata/National Council of Scientific and Technical Research of Argentina, La Plata, Argentina
| | - Giorgia Zadra
- Institute of Molecular Genetics, National Research Council, Pavia, Italy
| |
Collapse
|
21
|
He YL, Chen MT, Wang T, Zhang MM, Li YX, Wang HY, Ding N. Development of FABP4/5 inhibitors with potential therapeutic effect on type 2 Diabetes Mellitus. Eur J Med Chem 2021; 224:113720. [PMID: 34332399 DOI: 10.1016/j.ejmech.2021.113720] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/11/2021] [Accepted: 07/23/2021] [Indexed: 01/04/2023]
Abstract
Fatty acid-binding protein 4 (FABP4) and fatty acid-binding protein 5 (FABP5) are promising therapeutic targets for the treatment of various metabolic diseases. However, the weak potency, low selectivity over FABP3, or poor pharmacokinetic profiles of currently reported dual FABP4/5 inhibitors impeded further research. Here, we described the characterization of a series of dual FABP4/5 inhibitors with improved metabolic stabilities and physicochemical properties based on our previous studies. Among the compounds, D9 and E1 exhibited good inhibitory activities against FABP4/5 and favorable selectivity over FABP3 in vitro. In cell-based assays, D9 and E1 exerted a decrease of FABP4 secretion, a strong anti-lipolytic effect in mature adipocytes, and suppression of MCP-1 expression in THP-1 macrophages. Moreover, D9 and E1 possessed good metabolic stabilities in mouse hepatic microsomes and acceptable pharmacokinetics profiles in ICR mice. Further in vivo experiments showed that D9 and E1 could potently decrease serum FABP4 levels and ameliorate glucose metabolism disorders in obese diabetic db/db mice. These results demonstrated that D9 and E1 could serve as lead compounds for the development of novel anti-diabetic drugs.
Collapse
Affiliation(s)
- Yu-Long He
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Meng-Ting Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ting Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ming-Ming Zhang
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Ying-Xia Li
- School of Pharmacy, Fudan University, Shanghai, 201203, China.
| | - He-Yao Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Ning Ding
- School of Pharmacy, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
22
|
Fatty acids and evolving roles of their proteins in neurological, cardiovascular disorders and cancers. Prog Lipid Res 2021; 83:101116. [PMID: 34293403 DOI: 10.1016/j.plipres.2021.101116] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/04/2021] [Accepted: 07/14/2021] [Indexed: 01/03/2023]
Abstract
The dysregulation of fat metabolism is involved in various disorders, including neurodegenerative, cardiovascular, and cancers. The uptake of long-chain fatty acids (LCFAs) with 14 or more carbons plays a pivotal role in cellular metabolic homeostasis. Therefore, the uptake and metabolism of LCFAs must constantly be in tune with the cellular, metabolic, and structural requirements of cells. Many metabolic diseases are thought to be driven by the abnormal flow of fatty acids either from the dietary origin and/or released from adipose stores. Cellular uptake and intracellular trafficking of fatty acids are facilitated ubiquitously with unique combinations of fatty acid transport proteins and cytoplasmic fatty acid-binding proteins in every tissue. Extensive data are emerging on the defective transporters and metabolism of LCFAs and their clinical implications. Uptake and metabolism of LCFAs are crucial for the brain's functional development and cardiovascular health and maintenance. In addition, data suggest fatty acid metabolic transporter can normalize activated inflammatory response by reprogramming lipid metabolism in cancers. Here we review the current understanding of how LCFAs and their proteins contribute to the pathophysiology of three crucial diseases and the mechanisms involved in the processes.
Collapse
|
23
|
Liu Z, Xuan B, Tang S, Qian Z. Histone Deacetylase Inhibitor SAHA Induces Expression of Fatty Acid-Binding Protein 4 and Inhibits Replication of Human Cytomegalovirus. Virol Sin 2021; 36:1352-1362. [PMID: 34156645 DOI: 10.1007/s12250-021-00382-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 03/11/2021] [Indexed: 10/21/2022] Open
Abstract
Suberoylanilide hydroxamic acid (SAHA) is a histone deacetylase inhibitor that shows marked efficacy against many types of cancers and is approved to treat severe metastatic cutaneous T-cell lymphomas. In addition to its anticancer activity, SAHA has significant effects on the growth of many viruses. The effect of SAHA on replication of human cytomegalovirus (HCMV) has not, however, been investigated. Here, we showed that the replication of HCMV was significantly suppressed by treatment with SAHA at concentrations that did not show appreciable cytotoxicity. SAHA reduced transcription and protein levels of HCMV immediate early genes, showing that SAHA acts at an early stage in the viral life-cycle. RNA-sequencing data mining showed that numerous pathways and molecules were affected by SAHA. Interferon-mediated immunity was one of the most relevant pathways in the RNA-sequencing data, and we confirmed that SAHA inhibits HCMV-induced IFN-mediated immune responses using quantitative Real-time PCR (qRT-PCR). Fatty acid-binding protein 4 (FABP4), which plays a role in lipid metabolism, was identified by RNA-sequencing. We found that FABP4 expression was reduced by HCMV infection but increased by treatment with SAHA. We then showed that knockdown of FABP4 partially rescued the effect of SAHA on HCMV replication. Our data suggest that FABP4 contributes to the inhibitory effect of SAHA on HCMV replication.
Collapse
Affiliation(s)
- Zhongshun Liu
- CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Baoqin Xuan
- CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shubing Tang
- CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhikang Qian
- CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
24
|
Yin YY, Zhao J, Zhang LL, Xu XY, Liu JQ. Molecular mechanisms of inhibitor bindings to A-FABP deciphered by using molecular dynamics simulations and calculations of MM-GBSA. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2021; 32:293-315. [PMID: 33655818 DOI: 10.1080/1062936x.2021.1891966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/15/2021] [Indexed: 06/12/2023]
Abstract
Adipocyte fatty-acid binding protein (A-FABP) plays a central role in many aspects of metabolic diseases. It is an important target in drug design for treatment of FABP-related diseases. In this study, molecular dynamics (MD) simulations followed by calculations of molecular mechanics generalized Born surface area (MM-GBSA) and principal components analysis (PCA) were implemented to decipher molecular mechanism correlating with binding of inhibitors 57Q, 57P and L96 to A-FABP. The results show that van der Waals interactions are the leading factors to control associations of 57Q, 57P, and L96 with A-FABP, which reveals an energetic basis for designing of clinically available inhibitors towards A-FABP. The information from PCA and cross-correlation analysis rationally unveils that inhibitor bindings affect conformational changes of A-FABP and change relative movements between residues. Decomposition of binding affinity into contributions of individual residues not only detects hot spots of inhibitor/A-FABP binding but also shows that polar interactions of the positively charged residue Arg126 with three inhibitors provide a significant contribution for stabilization of the inhibitor/A-FABP bindings. Furthermore, the binding strength of L96 to residues Ser55, Phe57 and Lys58 are stronger than that of inhibitors 57Q and 57P to these residues.
Collapse
Affiliation(s)
- Y Y Yin
- School of Science, Shandong Jiaotong University, Jinan, China
| | - J Zhao
- School of Science, Shandong Jiaotong University, Jinan, China
| | - L L Zhang
- School of Science, Shandong Jiaotong University, Jinan, China
| | - X Y Xu
- School of Science, Shandong Jiaotong University, Jinan, China
| | - J Q Liu
- School of Science, Shandong Jiaotong University, Jinan, China
| |
Collapse
|
25
|
Jin X, Hao Z, Zhao M, Shen J, Ke N, Song Y, Qiao L, Lu Y, Hu L, Wu X, Wang J, Luo Y. MicroRNA-148a Regulates the Proliferation and Differentiation of Ovine Preadipocytes by Targeting PTEN. Animals (Basel) 2021; 11:ani11030820. [PMID: 33803986 PMCID: PMC7998426 DOI: 10.3390/ani11030820] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) have been found to be involved in lipid deposition and metabolism. However, there have been no reports on the roles of miR-148a in the proliferation and adipogenesis of preadipocytes in sheep. In this study, the expression of miR-148a was profiled in the eight tissues of Tibetan ewes and differentiated preadipocytes, and the role of miR-148a in differentiation and proliferation of ovine preadipocytes was investigated using Oil Red O staining, CCK-8, EdU staining, cell cycle detection, and RT-qPCR. The effect of PTEN on the differentiation of ovine preadipocytes was also investigated. The miR-148a was widely expressed in the eight tissues investigated and had significantly increased expression in liver, spleen and subcutaneous adipose tissues, and the heart. The expression of miR-148a continued to increase with the differentiation of ovine preadipocytes. The over-expression of miR-148a significantly promoted differentiation but inhibited the proliferation of ovine preadipocytes. The inhibition of miR-148a had the opposite effect on the differentiation and proliferation of ovine preadipocytes with over-expressed miR-148a. The results from the dual luciferase reporter assays showed that miR-148a mimic significantly decreased the luciferase activity of PTEN-3'UTR dual luciferase reporter vector, suggesting that PTEN is a target gene of miR-148a. In over-expressed-PTEN preadipocytes, the number of lipid droplets remarkably decreased, and the expression levels of adipogenesis marker genes PPARγ, FASN, FATP4, GLUT4, C/EBPβ and LPL were also significantly down-regulated. These results suggest that miR-148a accelerated the adipogenic differentiation of ovine preadipocytes by inhibiting PTEN expression, and also inhibited the proliferation of ovine preadipocytes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jiqing Wang
- Correspondence: (J.W.); (Y.L.); Tel.: +86-931-763-2469 (J.W.); +86-931-763-2483 (Y.L.)
| | - Yuzhu Luo
- Correspondence: (J.W.); (Y.L.); Tel.: +86-931-763-2469 (J.W.); +86-931-763-2483 (Y.L.)
| |
Collapse
|
26
|
Romanelli SM, MacDougald OA. Viral and Nonviral Transfer of Genetic Materials to Adipose Tissues: Toward a Gold Standard Approach. Diabetes 2020; 69:2581-2588. [PMID: 33219099 PMCID: PMC7679771 DOI: 10.2337/dbi20-0036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/04/2020] [Indexed: 01/03/2023]
Abstract
Gene transfer using viral or nonviral vectors enables the ability to manipulate specific cells and tissues for gene silencing, protein overexpression, or genome modification. Despite the widespread application of viral- and non-viral-mediated gene transfer to liver, heart, skeletal muscle, and the central nervous system, its use in adipose tissue has been limited. This is largely because adipose tissue is distributed throughout the body in distinct depots and adipocytes make up a minority of the cells within the tissue, making transduction difficult. Currently, there is no consensus methodology for efficient gene transfer to adipose tissue and many studies report conflicting information with regard to transduction efficiency and vector biodistribution. In this review, we summarize the challenges associated with gene transfer to adipose tissue and report on innovations that improve efficacy. We describe how vector and route of administration are the two key factors that influence transduction efficiency and outline a "gold standard" approach and experimental workflow for validating gene transfer to adipose tissue. Lastly, we speculate on how CRISPR/Cas9 can be integrated to improve adipose tissue research.
Collapse
Affiliation(s)
- Steven M Romanelli
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
| | - Ormond A MacDougald
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
27
|
Nguyen HC, Qadura M, Singh KK. Role of the Fatty Acid Binding Proteins in Cardiovascular Diseases: A Systematic Review. J Clin Med 2020; 9:E3390. [PMID: 33105856 PMCID: PMC7690604 DOI: 10.3390/jcm9113390] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) remain a global pandemic and leading cause of deaths worldwide. While several guidelines have been developed to control the development of CVDs, its prevalence keeps on increasing until this day. Cardiovascular risk factors, such as reduced exercises and high fat or glucose diets, culminate in the development of the metabolic syndrome and eventually atherosclerosis, which is driven by high blood lipid and cholesterol levels, and by endothelial dysfunction. Late complications of atherosclerosis give rise to serious clinical cardiovascular manifestations such as myocardial infarction and hypertension. Therefore, endothelial functions and the lipid metabolism play critical roles in the pathogenesis of CVDs. Fatty acid-binding proteins are a family of intracellular proteins expressed in many cell types known mainly for their interaction with and trafficking of cellular lipids. The roles of a number of isoforms in this family have been implicated in lipid metabolic homeostasis, but their influence on endothelial function and vascular homeostasis remain largely unknown. This review's purpose is to update fundamentals about the connection between cardiovascular disease, metabolism, endothelial function, and mainly the roles of fatty acid-binding proteins.
Collapse
Affiliation(s)
- Hien C. Nguyen
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada;
| | - Mohammad Qadura
- Vascular Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada;
| | - Krishna K. Singh
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada;
- Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
- Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
28
|
Kwong SC, Abd Jamil AH, Rhodes A, Taib NA, Chung I. Fatty acid binding protein 7 mediates linoleic acid-induced cell death in triple negative breast cancer cells by modulating 13-HODE. Biochimie 2020; 179:23-31. [PMID: 32931863 DOI: 10.1016/j.biochi.2020.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 08/11/2020] [Accepted: 09/07/2020] [Indexed: 01/01/2023]
Abstract
Different fatty acids have distinct effects on the survival of breast cancer cells, which could be mediated by fatty acid binding proteins (FABPs), a family of lipid chaperones. Due to the diverse structures of the members of FABP family, each FABP demonstrates distinct binding affinities to different fatty acids. Of note, FABP7 is predominantly expressed in triple negative breast cancer (TNBC), the most aggressive subtype of breast cancer. Yet, the role of FABP7 in modulating the effects of fatty acids on TNBC survival was unclear. In contrast to the high expression of FABP7 in human TNBC tumours, FABP7 protein was undetectable in TNBC cell lines. Hence, a FABP7 overexpression model was used for this study, in which the transduced TNBC cell lines (MDA-MB-231 and Hs578T) were treated with various mono- and polyunsaturated fatty acids. Oleic acid (OA), docosahexaenoic acid (DHA) and arachidonic acid (AA) inhibited TNBC cell growth at high concentrations, with no differences resulted from FABP7 overexpression. Interestingly, overexpression of FABP7 augmented linoleic acid-induced cell death in MDA-MB-231 cells. The increased cell death may be explained by a decrease in 13-HODE, a pro-tumorigenic oxidation product of linoleic acid. The phenotype was, however, attenuated with a rescue treatment using 25 nM 13-HODE. The decrease in 13-HODE was potentially due to fatty acid partitioning modulated by FABP7, as demonstrated by a 3-fold increase in fatty acid oxidation. Our findings suggest that linoleic acid could be a potential therapeutic strategy for FABP7-overexpressing TNBC patients.
Collapse
Affiliation(s)
- Soke Chee Kwong
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Amira Hajirah Abd Jamil
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Anthony Rhodes
- Department of Pathology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia; School of Health Sciences, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Nur Aishah Taib
- Department of Surgery, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia; University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Ivy Chung
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia; University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
29
|
Mechanisms linking adipose tissue inflammation to cardiac hypertrophy and fibrosis. Clin Sci (Lond) 2020; 133:2329-2344. [PMID: 31777927 DOI: 10.1042/cs20190578] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/08/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022]
Abstract
Adipose tissue is classically recognized as the primary site of lipid storage, but in recent years has garnered appreciation for its broad role as an endocrine organ comprising multiple cell types whose collective secretome, termed as adipokines, is highly interdependent on metabolic homeostasis and inflammatory state. Anatomical location (e.g. visceral, subcutaneous, epicardial etc) and cellular composition of adipose tissue (e.g. white, beige, and brown adipocytes, macrophages etc.) also plays a critical role in determining its response to metabolic state, the resulting secretome, and its potential impact on remote tissues. Compared with other tissues, the heart has an extremely high and constant demand for energy generation, of which most is derived from oxidation of fatty acids. Availability of this fatty acid fuel source is dependent on adipose tissue, but evidence is mounting that adipose tissue plays a much broader role in cardiovascular physiology. In this review, we discuss the impact of the brown, subcutaneous, and visceral white, perivascular (PVAT), and epicardial adipose tissue (EAT) secretome on the development and progression of cardiovascular disease (CVD), with a particular focus on cardiac hypertrophy and fibrosis.
Collapse
|
30
|
Comparison between Tibetan and Small-tailed Han sheep in adipocyte phenotype, lipid metabolism and energy homoeostasis regulation of adipose tissues when consuming diets of different energy levels. Br J Nutr 2020; 124:668-680. [PMID: 32406340 DOI: 10.1017/s0007114520001701] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
This study aimed to gain insight into how adipose tissue of Tibetan sheep regulates energy homoeostasis to cope with low energy intake under the harsh environment of the Qinghai-Tibetan Plateau (QTP). We compared Tibetan and Small-tailed Han sheep (n 24 of each breed), all wethers and 1·5 years of age, which were each divided randomly into four groups and offered diets of different digestible energy (DE) densities: 8·21, 9·33, 10·45 and 11·57 MJ DE/kg DM. When the sheep lost body mass and were assumed to be in negative energy balance: (1) adipocyte diameter in subcutaneous adipose tissue was smaller and decreased to a greater extent in Tibetan than in Small-tailed Han sheep, but the opposite occurred in the visceral adipose tissue; (2) Tibetan sheep showed higher insulin receptor mRNA expression and lower concentrations of catabolic hormones than Small-tailed Han sheep and (3) Tibetan sheep had lower capacity for glucose and fatty acid uptake than Small-tailed Han sheep. Moreover, Tibetan sheep had lower AMPKα mRNA expression but higher mammalian target of rapamycin mRNA expression in the adipocytes than Small-tailed Han sheep. We concluded that Tibetan sheep had lower catabolism but higher anabolism in adipose tissue and reduced the capacity for glucose and fatty acid uptake to a greater extent than Small-tailed Han sheep to maintain energy homoeostasis when in negative energy balance. These responses provide Tibetan sheep with a high ability to cope with low energy intake and with the harsh environment of the QTP.
Collapse
|
31
|
Hofer P, Taschler U, Schreiber R, Kotzbeck P, Schoiswohl G. The Lipolysome-A Highly Complex and Dynamic Protein Network Orchestrating Cytoplasmic Triacylglycerol Degradation. Metabolites 2020; 10:E147. [PMID: 32290093 PMCID: PMC7240967 DOI: 10.3390/metabo10040147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/03/2020] [Accepted: 04/08/2020] [Indexed: 12/25/2022] Open
Abstract
The catabolism of intracellular triacylglycerols (TAGs) involves the activity of cytoplasmic and lysosomal enzymes. Cytoplasmic TAG hydrolysis, commonly termed lipolysis, is catalyzed by the sequential action of three major hydrolases, namely adipose triglyceride lipase, hormone-sensitive lipase, and monoacylglycerol lipase. All three enzymes interact with numerous protein binding partners that modulate their activity, cellular localization, or stability. Deficiencies of these auxiliary proteins can lead to derangements in neutral lipid metabolism and energy homeostasis. In this review, we summarize the composition and the dynamics of the complex lipolytic machinery we like to call "lipolysome".
Collapse
Affiliation(s)
- Peter Hofer
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; (P.H.); (U.T.); (R.S.)
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; (P.H.); (U.T.); (R.S.)
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; (P.H.); (U.T.); (R.S.)
| | - Petra Kotzbeck
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria;
| | - Gabriele Schoiswohl
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; (P.H.); (U.T.); (R.S.)
| |
Collapse
|
32
|
Su H, Zou Y, Chen G, Dou H, Xie H, Yuan X, Zhang X, Zhang N, Li M, Xu Y. Exploration of Fragment Binding Poses Leading to Efficient Discovery of Highly Potent and Orally Effective Inhibitors of FABP4 for Anti-inflammation. J Med Chem 2020; 63:4090-4106. [DOI: 10.1021/acs.jmedchem.9b02107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Haixia Su
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Zou
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guofeng Chen
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huixia Dou
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hang Xie
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaojing Yuan
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xianglei Zhang
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Naixia Zhang
- Department of Analytical Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minjun Li
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yechun Xu
- CAS Key Laboratory of Receptor Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
33
|
Yilmaz AB, Tapsin S, Elbasan EB, Kayhan HD, Sahin F, Turkel N. Suppressor Effects of Sodium Pentaborate Pentahydrate and Pluronic F68 on Adipogenic Differentiation and Fat Accumulation. Biol Trace Elem Res 2020; 193:390-399. [PMID: 31119640 DOI: 10.1007/s12011-019-01738-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/17/2019] [Indexed: 10/26/2022]
Abstract
Obesity is a major public health problem worldwide and a risk factor for certain diseases, including cardiovascular disease, diabetes, cancer, and depression. Unfortunately, currently available anti-obesity drugs have failed in the long-term maintenance of weight control. It has been a challenge to design novel drugs that could potentially treat obesity or prevent uncontrolled weight-gain which lies underneath the pathology of obesity. Since obesity in a way is a consequence of the accumulating new mature adipocytes from undifferentiated precursors which is a process also termed as adipogenesis, drugs that might control adipogenesis could be beneficial for the treatment of obesity. In the current study, combined effect of sodium pentaborate pentahydrate (NaB) and pluronic F68 on adipogenic differentiation was examined by administering various combinations of the two agents to human adipose-derived stem cells (hADSCs) in in vitro. Immunocytochemistry and quantitative RT-PCR were performed to evaluate the levels of adipogenesis-promoting genes such as peroxisome proliferator-activated receptor-γ (PPARγ), fatty acid binding protein (FABP4), and adiponectin. Results indicated that expressions of all these three genes were restrained. Furthermore, Oil Red O staining revealed that lipid vesicle formation was reduced in hADSCs treated with differentiation medium containing NaB/F68 combination. Finally, expression levels of Hippo pathway kinases Lats2, MST1, and scaffold protein Sav1 were reduced in these cells, suggesting a possible link between Hippo pathway-dependent downregulation of PPARγ and the NaB/F68 treatment. Herein, we showed that combination of NaB and F68 curtails adipocyte differentiation by inhibiting the adipogenic transcriptional program leading to a decrease in lipid accumulation in adipocytes even at very low doses, thereby uncovered a striking opportunity to use this combination in obesity treatment.
Collapse
Affiliation(s)
- Aysu Bilge Yilmaz
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Kayışdağı, 34755, Istanbul, Turkey
- Koc Universitesi Hastanesi, Davutpasa Cd No:4, Topkapi Zeytinburnu, 34090, Istanbul, Turkey
| | - Sidika Tapsin
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Kayışdağı, 34755, Istanbul, Turkey
- Stem Cell and Development Biology, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Elif Burce Elbasan
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Kayışdağı, 34755, Istanbul, Turkey
| | - Hatice Damla Kayhan
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Kayışdağı, 34755, Istanbul, Turkey
- Yaşam Bilimleri ve Teknolojileri Uygulama ve Araştırma Merkezi, Boğaziçi Üniversitesi, Kuzey Kampüs ETA-B Blok. 4.Kat, 34342, Bebek/Istanbul, Turkey
| | - Fikrettin Sahin
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Kayışdağı, 34755, Istanbul, Turkey
| | - Nezaket Turkel
- Faculty of Engineering, Department of Genetics and Bioengineering, Yeditepe University, Kayışdağı, 34755, Istanbul, Turkey.
| |
Collapse
|
34
|
HWANG JS, LEE SB, CHOI MJ, KIM JT, SEO HG. Anti-adipogenic effect of a turmeric extract-loaded nanoemulsion in 3T3-L1 preadipocytes and high fat diet-fed mice. FOOD SCIENCE AND TECHNOLOGY 2019. [DOI: 10.1590/fst.20718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
35
|
Li B, Qiao L, Yan X, Shi T, Ren D, Zhao Y, Zhao J, Liu W. mRNA expression of genes related to fat deposition during in vitro ovine adipogenesis. CANADIAN JOURNAL OF ANIMAL SCIENCE 2019. [DOI: 10.1139/cjas-2018-0107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Fat deposition in animals involves adipogenic differentiation guided by transcriptional factors and other key factors. To understand the molecular mechanism underlying ovine adipogenic differentiation, the dynamic mRNA expression of key genes related to fat deposition, including peroxisome proliferator-activated receptor-γ (PPAR-γ), fatty acid-binding protein 4 (FABP4), FABP5, and cellular retinoic acid-binding protein 2 (CRABP2), were analyzed during in vitro differentiation of ovine preadipocytes. The stromal vascular cells from underneath the tail fat tissue of 1-wk-old sheep were isolated and cultured, and the preadipocytes were induced using a cocktail of 3-isobutyl-1-methylxanthine, insulin, dexamethasone, and troglitazone. The cultivated cells were collected at different time points after induced differentiation. The expression levels of PPAR-γ, FABP4, FABP5, and CRABP2 were studied by quantitative real-time polymerase chain reaction. The expressions of these genes in sheep were compared with those in human and mouse retrieved from the Gene Expression Omnibus DataSets. We observed that the expression of PPAR-γ, FABP4, and FABP5 was increased upon differentiation of ovine preadipocytes, as in humans and mice. The expression of CRABP2 was sharply increased from days 0 to 2 after induced differentiation and was subsequently decreased. This expression pattern of CRABP2 was different from that observed in humans and mice. Our results provide new insights into the function of these genes in fat deposition.
Collapse
Affiliation(s)
- Baojun Li
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Liying Qiao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Xiaoru Yan
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Tao Shi
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Duanyang Ren
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Yanyan Zhao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Junxing Zhao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| | - Wenzhong Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, People’s Republic of China
| |
Collapse
|
36
|
Pomar CA, Kuda O, Kopecky J, Rombaldova M, Castro H, Picó C, Sánchez J, Palou A. Maternal diet, rather than obesity itself, has a main influence on milk triacylglycerol profile in dietary obese rats. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1865:158556. [PMID: 31678620 DOI: 10.1016/j.bbalip.2019.158556] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 01/31/2023]
Abstract
Triacylglycerols (TG) in milk derive from different sources, and their composition may be influenced by both maternal diet and obesity. We used two rat models to ascertain potential changes in TG composition in milk associated to maternal intake of an obesogenic diet during lactation and to distinguish them from the effects attributable to maternal adiposity. Milk samples were obtained from dams fed a cafeteria diet during lactation (CAF) and from dams made obese by cafeteria diet feeding, with dietary normalization before gestation (PCaf). Levels of specific TG species in milk collected at different time points of lactation were determined by shotgun lipidomics. CAF and PCaf dams presented a greater adiposity than their respective controls. The principal component analysis of TG peaks showed a clear separation between milk from CAF dams and milk from control and Pcaf dams, already evident at 5 days of lactation. Milk from CAF dams was enriched with TG species with greater number of carbons and double bonds and reduced in TG with lower number of carbons. TG composition of milk from Pcaf dams was similar to controls, although specific differences were observed at day 5 of lactation. Thus, the intake of a cafeteria diet during lactation, rather than maternal adiposity, alters milk composition. This effect is avoided with dietary normalization before gestation, although the remaining fat reserves may also influence TG composition at initial stages of lactation. Therefore, normalization of maternal diet prior to pregnancy should be considered as a strategy for achieving optimal milk composition.
Collapse
Affiliation(s)
- C A Pomar
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics and Obesity), Palma de Mallorca, Spain; University of the Balearic Islands and CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Palma de Mallorca, Spain; Instituto de Investigación Sanitaria Illes Balears, Palma de Mallorca, Spain
| | - O Kuda
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - J Kopecky
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - M Rombaldova
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - H Castro
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics and Obesity), Palma de Mallorca, Spain; Universidad Autónoma de Nuevo León, Facultad de Salud Pública y Nutrición, Nuevo León, Mexico
| | - C Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics and Obesity), Palma de Mallorca, Spain; University of the Balearic Islands and CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Palma de Mallorca, Spain; Instituto de Investigación Sanitaria Illes Balears, Palma de Mallorca, Spain
| | - J Sánchez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics and Obesity), Palma de Mallorca, Spain; University of the Balearic Islands and CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Palma de Mallorca, Spain; Instituto de Investigación Sanitaria Illes Balears, Palma de Mallorca, Spain.
| | - A Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics and Obesity), Palma de Mallorca, Spain; University of the Balearic Islands and CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Palma de Mallorca, Spain; Instituto de Investigación Sanitaria Illes Balears, Palma de Mallorca, Spain
| |
Collapse
|
37
|
Cohen-Zinder M, Lipkin E, Shor-Shimoni E, Ben-Meir Y, Agmon R, Asher A, Miron J, Shabtay A. FABP4 gene has a very large effect on feed efficiency in lactating Israeli Holstein cows. Physiol Genomics 2019; 51:481-487. [PMID: 31373885 DOI: 10.1152/physiolgenomics.00051.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Improving feed efficiency (FE) is a major goal for the livestock industry. Previously, we have identified 48 SNP markers distributed over 32 genes significantly associated with residual feed intake (RFI) in Israeli Holstein male calves, the most significant of which are located in the bovine FABP4 gene. In the present study, we tested associations of eight of the FABP4 markers with RFI and feed conversion ratio (FCR), along with milk composition and feeding behavioral traits, in 114 lactating Israeli Holstein cows. Large allele effects were found, along with large contributions of FABP4 markers to the phenotypic variation [mean contribution of all significant markers (P < 0.05), 15.4 and 12.0% for RFI and FCR, respectively] and genotypic variation [means of all significant markers (P < 0.05), 75.7 and 32.4% in RFI and FCR, respectively]. However, the association of all significant FABP4 markers with FE and milk content traits was found in opposite directions, such that improved FE was accompanied by decreased milk content. Hence, before inclusion in breeding programs, the gain in FE must be economically balanced with the loss in milk contents. On the other hand, these findings imply that in any current improvement program concentrated on milk traits alone, without taking into account the effect on FE, the progress in milk composition is probably accompanied by deterioration of FE. These results, if confirmed in other populations and breeds, set FABP4 as a prime candidate in any marker-assisted selection program targeting FE as a whole and RFI in particular.
Collapse
Affiliation(s)
- Miri Cohen-Zinder
- Beef Cattle Section, Newe-Ya'ar Research Center, Agricultural Research Organization, Ramat Yishay, 30095, Israel
| | - Ehud Lipkin
- Department of Genetics, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Einav Shor-Shimoni
- Beef Cattle Section, Newe-Ya'ar Research Center, Agricultural Research Organization, Ramat Yishay, 30095, Israel
| | - Yehoshav Ben-Meir
- Department of Ruminant Sciences, Agricultural Research Organization, Bet Dagan, Israel
| | - Rotem Agmon
- Beef Cattle Section, Newe-Ya'ar Research Center, Agricultural Research Organization, Ramat Yishay, 30095, Israel
| | - Aviv Asher
- Northern R&D, MIGAL, Galilee Technology Center, Kiryat Shmona, Israel
| | - Joshua Miron
- Department of Ruminant Sciences, Agricultural Research Organization, Bet Dagan, Israel
| | - Ariel Shabtay
- Beef Cattle Section, Newe-Ya'ar Research Center, Agricultural Research Organization, Ramat Yishay, 30095, Israel
| |
Collapse
|
38
|
Identification of new dual FABP4/5 inhibitors based on a naphthalene-1-sulfonamide FABP4 inhibitor. Bioorg Med Chem 2019; 27:115015. [PMID: 31420256 DOI: 10.1016/j.bmc.2019.07.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 12/30/2022]
Abstract
Fatty acid binding protein 4 (FABP4) and fatty acid binding protein 5 (FABP5) are mainly expressed in adipocytes and/or macrophages and play essential roles in energy metabolism and inflammation. When FABP4 function is diminished, FABP5 expression is highly increased possibly as a functional compensation. Dual FABP4/5 inhibitors are expected to provide beneficial synergistic effect on treating diabetes, atherosclerosis, and inflammation-related diseases. Starting from our previously reported selective FABP4 inhibitor 8, structural biology information was used to modulate the selectivity profile and to design potent dual FABP4/5 inhibitors with good selectivity against FABP3. Two compounds A16 and B8 were identified to show inhibitory activities against both FABP4/5 and good selectivity over FABP3, which could also reduce the level of forskolin-stimulated lipolysis in mature 3T3-L1 adipocytes. Compared with compound 8, these two compounds exhibited better anti-inflammatory effects in lipopolysaccharide-stimulated RAW264.7 murine macrophages, with decreased levels of pro-inflammatory cytokines TNFα and MCP-1 and apparently inhibited IKK/NF-κB pathway.
Collapse
|
39
|
Kwong SC, Jamil AHA, Rhodes A, Taib NA, Chung I. Metabolic role of fatty acid binding protein 7 in mediating triple-negative breast cancer cell death via PPAR-α signaling. J Lipid Res 2019; 60:1807-1817. [PMID: 31484694 DOI: 10.1194/jlr.m092379] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 08/06/2019] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, partly due to the lack of targeted therapy available. Cancer cells heavily reprogram their metabolism and acquire metabolic plasticity to satisfy the high-energy demand due to uncontrolled proliferation. Accumulating evidence shows that deregulated lipid metabolism affects cancer cell survival, and therefore we sought to understand the function of fatty acid binding protein 7 (FABP7), which is expressed predominantly in TNBC tissues. As FABP7 was not detected in the TNBC cell lines tested, Hs578T and MDA-MB-231 cells were transduced with lentiviral particles containing either FABP7 open reading frame or red fluorescent protein. During serum starvation, when lipids were significantly reduced, FABP7 decreased the viability of Hs578T, but not of MDA-MB-231, cells. FABP7-overexpressing Hs578T (Hs-FABP7) cells failed to efficiently utilize other available bioenergetic substrates such as glucose to sustain ATP production, which led to S/G2 phase arrest and cell death. We further showed that this metabolic phenotype was mediated by PPAR-α signaling, despite the lack of fatty acids in culture media, as Hs-FABP7 cells attempted to survive. This study provides imperative evidence of metabolic vulnerabilities driven by FABP7 via PPAR-α signaling.
Collapse
Affiliation(s)
- Soke Chee Kwong
- Departments of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Anthony Rhodes
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Lakeside Campus, 47500 Subang Jaya, Selangor, Malaysia.,Pathology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Nur Aishah Taib
- Surgery Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.,University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Ivy Chung
- Departments of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia .,University of Malaya Cancer Research Institute, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
40
|
Josephrajan A, Hertzel AV, Bohm EK, McBurney MW, Imai SI, Mashek DG, Kim DH, Bernlohr DA. Unconventional Secretion of Adipocyte Fatty Acid Binding Protein 4 Is Mediated By Autophagic Proteins in a Sirtuin-1-Dependent Manner. Diabetes 2019; 68:1767-1777. [PMID: 31171562 PMCID: PMC6702637 DOI: 10.2337/db18-1367] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Fatty acid binding protein 4 (FABP4) is a leaderless lipid carrier protein primarily expressed by adipocytes and macrophages that not only functions intracellularly but is also secreted. The secretion is mediated via unconventional mechanism(s), and in a variety of species, metabolic dysfunction is correlated with elevated circulating FABP4 levels. In diabetic animals, neutralizing antibodies targeting serum FABP4 increase insulin sensitivity and attenuate hepatic glucose output, suggesting the functional importance of circulating FABP4. Using animal and cell-based models, we show that FABP4 is secreted from white, but not brown, adipose tissue in response to lipolytic stimulation in a sirtuin-1 (SIRT1)-dependent manner via a mechanism that requires some, but not all, autophagic components. Silencing of early autophagic genes such as Ulk1/2, Fip200, or Beclin-1 or chemical inhibition of ULK1/2 or VPS34 attenuated secretion, while Atg5 knockdown potentiated FABP4 release. Genetic knockout of Sirt1 diminished secretion, and serum FABP4 levels were undetectable in Sirt1 knockout mice. In addition, blocking SIRT1 by EX527 attenuated secretion while activating SIRT1 by resveratrol-potentiated secretion. These studies suggest that FABP4 secretion from adipocytes is regulated by SIRT1 and requires early autophagic components.
Collapse
Affiliation(s)
- Ajeetha Josephrajan
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Ann V Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Ellie K Bohm
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Michael W McBurney
- Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Ottawa, Ontario, Canada
| | - Shin-Ichiro Imai
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Do-Hyung Kim
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
41
|
The Effects of Radix Astragali Water Abstract on Energy Metabolism in Rat Yang-Deficiency Cold Syndrome Model through PPAR Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:9194362. [PMID: 30519272 PMCID: PMC6241352 DOI: 10.1155/2018/9194362] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/09/2018] [Indexed: 12/23/2022]
Abstract
Radix Astragali (RA) herb with warm property and significant “tonifying qi” effects is indicated for the syndrome of internal cold due to Yang deficiency. The purpose of this research was to explore effects of Radix Astragali (RA) through PPAR signaling pathway on gene expression profiles related to energy metabolism in rats with the Yang-deficiency cold (YDC) syndrome, for identifying the pathological mechanism of Yang-deficiency cold (YDC) syndrome and the effects mechanism of RA. The results indicated that RA could significantly increase body weight (BM), cold and heat tendency (CT), overall temperature (OT), rectum temperature (RT), toe temperature (TT), energy intake (EI), and V(O2)/V(CO2) ratio (which indicates basal metabolism, BM) (P<0.05), enhancing the depressed metabolic function in YDC syndrome model rat. Our data also indicated differentially expressed genes (DEGs) related to energy metabolism involving lipids, carbohydrates, and amino acids metabolic process; the expression of CPT-1 and FABP4 (ap2) was improved; PPAR, Glycolysis, Wnt, cAMP, MAPK, AMPK, and fatty acid degradation signaling pathway may be related to energy metabolism. However, the Chinese herbal medicine RA plays a certain role in promoting the metabolism of substances and energy in rats by its warming and beneficial effect. Our results suggest that the mechanism underlying the function of RA may take effect through the regulation of PPAR signaling pathway and related gene expression. Lipids, carbohydrates, and amino acids metabolic process may be affected to adjust the reduced metabolic function in the model animals. In general, results indicate that RA could promote energy metabolism in rats with the YDC syndrome via PPAR signaling pathway regulating the expression of CPT-1 and FABP4 (ap2), which reflected the warm and qi tonifying properties of RA.
Collapse
|
42
|
Ge XN, Bastan I, Dileepan M, Greenberg Y, Ha SG, Steen KA, Bernlohr DA, Rao SP, Sriramarao P. FABP4 regulates eosinophil recruitment and activation in allergic airway inflammation. Am J Physiol Lung Cell Mol Physiol 2018; 315:L227-L240. [PMID: 29696987 PMCID: PMC6139653 DOI: 10.1152/ajplung.00429.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 04/17/2018] [Accepted: 04/18/2018] [Indexed: 12/11/2022] Open
Abstract
Fatty acid binding protein 4 (FABP4), a member of a family of lipid-binding proteins, is known to play a role in inflammation by virtue of its ability to regulate intracellular events such as lipid fluxes and signaling. Studies have indicated a proinflammatory role for FABP4 in allergic asthma although its expression and function in eosinophils, the predominant inflammatory cells recruited to allergic airways, were not investigated. We examined expression of FABP4 in murine eosinophils and its role in regulating cell recruitment in vitro as well as in cockroach antigen (CRA)-induced allergic airway inflammation. CRA exposure led to airway recruitment of FABP4-expressing inflammatory cells, specifically eosinophils, in wild-type (WT) mice. FABP4 expression in eosinophils was induced by TNF-α as well as IL-4 and IL-13. FABP4-deficient eosinophils exhibited markedly decreased cell spreading/formation of leading edges on vascular cell adhesion molecule-1 and significantly decreased adhesion to intercellular adhesion molecule-1 associated with reduced β2-integrin expression relative to WT cells. Furthermore, FABP4-deficient eosinophils exhibited decreased migration, F-actin polymerization, calcium flux, and ERK(1/2) phosphorylation in response to eotaxin-1. In vivo, CRA-challenged FABP4-deficient mice exhibited attenuated eosinophilia and significantly reduced airway inflammation (improved airway reactivity, lower IL-5, IL-13, TNF-α, and cysteinyl leukotriene C4 levels, decreased airway structural changes) compared with WT mice. In conclusion, expression of FABP4 in eosinophils is induced during conditions of inflammation and plays a proinflammatory role in the development of allergic asthma by promoting eosinophil adhesion and migration and contributing to the development of various aspects of airway inflammation.
Collapse
Affiliation(s)
- Xiao Na Ge
- Laboratory of Allergic Diseases and Inflammation, Department of Veterinary and Biomedical Sciences, Saint Paul, Minnesota
| | - Idil Bastan
- Laboratory of Allergic Diseases and Inflammation, Department of Veterinary and Biomedical Sciences, Saint Paul, Minnesota
| | - Mythili Dileepan
- Laboratory of Allergic Diseases and Inflammation, Department of Veterinary and Biomedical Sciences, Saint Paul, Minnesota
| | - Yana Greenberg
- Laboratory of Allergic Diseases and Inflammation, Department of Veterinary and Biomedical Sciences, Saint Paul, Minnesota
| | - Sung Gil Ha
- Laboratory of Allergic Diseases and Inflammation, Department of Veterinary and Biomedical Sciences, Saint Paul, Minnesota
| | - Kaylee A. Steen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Saint Paul, Minnesota
| | - David A. Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Saint Paul, Minnesota
| | - Savita P. Rao
- Laboratory of Allergic Diseases and Inflammation, Department of Veterinary and Biomedical Sciences, Saint Paul, Minnesota
| | - P. Sriramarao
- Laboratory of Allergic Diseases and Inflammation, Department of Veterinary and Biomedical Sciences, Saint Paul, Minnesota
| |
Collapse
|
43
|
Zhang C, Chiu KY, Chan BPM, Li T, Wen C, Xu A, Yan CH. Knocking out or pharmaceutical inhibition of fatty acid binding protein 4 (FABP4) alleviates osteoarthritis induced by high-fat diet in mice. Osteoarthritis Cartilage 2018; 26:824-833. [PMID: 29549054 DOI: 10.1016/j.joca.2018.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 01/01/2018] [Accepted: 03/06/2018] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Adipokines play roles in the pathogenesis of osteoarthritis (OA). Fatty acid binding protein 4 (FABP4) is a novel adipokine that is closely associated with obesity and metabolic diseases. The aim of this study was to discover the potential role of FABP4 in OA. METHODS Seventy-two FABP4 knockout mice (KO) in C57BL/6N background and wild-type littermates (WT) (male, 6-week-old) were fed with a high-fat diet (HFD, 60% calorie) or standard diet (STD, 11.6% calorie) for 3 months, 6 months and 9 months (n = 6 each). In the parallel study, forty-eight 6-week-old male WT mice were fed with HFD or STD, and simultaneously treated with daily oral gavage of selective FABP4 inhibitor BMS309403 (15 mg/kg/d) or vehicle for 4 months and 6 months (n = 6 each). Serum FABP4 and cartilage oligomeric matrix protein (COMP) concentration was quantified. Histological assessment of knee OA and micro-CT analysis of subchondral bone were performed. RESULTS HFD induced obesity in mice. After 3 months and 6 months of HFD, KO mice showed alleviated cartilage degradation and synovitis, with significantly lower COMP, modified Mankin OA score, and MMP-13/ADAMTS4 expression. After 6 months and 9 months of HFD, KO mice showed less osteophyte formation and subchondral bone sclerosis. Chronic treatment of BMS309403 for 4 months and 6 months significantly alleviated cartilage degradation, but had no effects on the subchondral bone. Knocking out or pharmaceutical inhibition of FABP4 did not have significant effects on lean mice fed with STD. CONCLUSIONS Knocking out or pharmaceutical inhibition of FABP4 alleviates OA induced by HFD in mice.
Collapse
Affiliation(s)
- C Zhang
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong Special Administrative Region; Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
| | - K Y Chiu
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong Special Administrative Region.
| | - B P M Chan
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong Special Administrative Region.
| | - T Li
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong Special Administrative Region.
| | - C Wen
- Department of Biomedical Engineering, Faculty of Engineering, Hong Kong Polytechnic University, Hong Kong Special Administrative Region.
| | - A Xu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region.
| | - C H Yan
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong Special Administrative Region; Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
44
|
Gao DD, Dou HX, Su HX, Zhang MM, Wang T, Liu QF, Cai HY, Ding HP, Yang Z, Zhu WL, Xu YC, Wang HY, Li YX. From hit to lead: Structure-based discovery of naphthalene-1-sulfonamide derivatives as potent and selective inhibitors of fatty acid binding protein 4. Eur J Med Chem 2018; 154:44-59. [DOI: 10.1016/j.ejmech.2018.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/04/2018] [Accepted: 05/06/2018] [Indexed: 01/05/2023]
|
45
|
An SM, Seong KY, Yim SG, Hwang YJ, Bae SH, Yang SY, An BS. Intracutaneous delivery of gelatins induces lipolysis and suppresses lipogenesis of adipocytes. Acta Biomater 2018; 67:238-247. [PMID: 29208554 DOI: 10.1016/j.actbio.2017.11.050] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/24/2017] [Accepted: 11/28/2017] [Indexed: 12/21/2022]
Abstract
Due to growing interest in cosmetics and medical applications, therapeutic medications that reduce the amount of local subcutaneous adipose tissue have potential for obesity treatment. However, conventional methods such as surgical operation are restricted due to risk of complications. Here, we report a simple and effective method for local reduction of subcutaneous adipose tissue (AT) by using microneedle-assisted transdermal delivery of natural polymers. After in vitro screening tests, gelatin was selected as a therapeutic polymer to reduce accumulation of AT. An in vitro study showed that the level of released glycerol as an indicator of lipolysis was elevated in isolated adipocytes after gelatin treatment. In addition, gelatins suppressed expression levels of lipogenesis-associated genes. Following application of gelatin microneedle (GMN) patches to high-fat diet (HD)-induced obese rats, the amount of subcutaneous AT at the site of GMN application was significantly reduced, which was also confirmed by histological analysis and micro-computed tomography scanning. In addition, lipogenesis-associated genes were down-regulated in GMN-treated subcutaneous AT. These findings suggest that GMN patches induce lipolysis and simultaneously inhibit lipogenesis, thereby reducing deposition of subcutaneous AT. This platform using GMNs may provide a new strategy to treat excess subcutaneous AT with minimal complications. STATEMENT OF SIGNIFICANCE: (1) Significance This work reports a new approach for the local reduction of subcutaneous adipose tissue using a dissolving microneedle patch prepared using gelatin to enable suppression of lipogenesis and acceleration of lipolysis in adipocytes. The gelatin microneedle patch exhibited a significant reduction of local subcutaneous fat up to 60% compared to control groups without any change in total weight. (2) Scientific impact This is the first report demonstrating the direct anti-obesity effects of gelatin administrated in a transdermal route and the feasibility of natural polymer therapeutics for regional reduction of subcutaneous fat. We believe that our work will excite interdisciplinary readers of Acta Biomaterialia, those who are interested in the natural polymers, drug delivery, and obesity.
Collapse
Affiliation(s)
- Sung-Min An
- Department of Biomaterials Science, Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Keum-Yong Seong
- Department of Biomaterials Science, Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Sang-Gu Yim
- Department of Biomaterials Science, Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Young Jun Hwang
- Department of Biomaterials Science, Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Republic of Korea
| | - Seong Hwan Bae
- Department of Plastic and Reconstructive Surgery, Pusan National University, Busan 46241, Republic of Korea
| | - Seung Yun Yang
- Department of Biomaterials Science, Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Republic of Korea.
| | - Beum-Soo An
- Department of Biomaterials Science, Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Republic of Korea.
| |
Collapse
|
46
|
Walenna NF, Kurihara Y, Chou B, Ishii K, Soejima T, Itoh R, Shimizu A, Ichinohe T, Hiromatsu K. Chlamydia pneumoniae exploits adipocyte lipid chaperone FABP4 to facilitate fat mobilization and intracellular growth in murine adipocytes. Biochem Biophys Res Commun 2017; 495:353-359. [PMID: 29108997 DOI: 10.1016/j.bbrc.2017.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 11/02/2017] [Indexed: 11/28/2022]
Abstract
Fatty acid-binding protein 4 (FABP4), a cytosolic lipid chaperone predominantly expressed in adipocytes and macrophages, modulates lipid fluxes, trafficking, signaling, and metabolism. Recent studies have demonstrated that FABP4 regulates metabolic and inflammatory pathways, and in mouse models its inhibition can improve type 2 diabetes mellitus and atherosclerosis. However, the role of FABP4 in bacterial infection, metabolic crosstalk between host and pathogen, and bacterial pathogenesis have not been studied. As an obligate intracellular pathogen, Chlamydia pneumoniae needs to obtain nutrients such as ATP and lipids from host cells. Here, we show that C. pneumoniae successfully infects and proliferates in murine adipocytes by inducing hormone sensitive lipase (HSL)-mediated lipolysis. Chemical inhibition or genetic manipulation of HSL significantly abrogated the intracellular growth of C. pneumoniae in adipocytes. Liberated free fatty acids were utilized to generate ATP via β-oxidation, which C. pneumoniae usurped for its replication. Strikingly, chemical inhibition or genetic silencing of FABP4 significantly abrogated C. pneumoniae infection-induced lipolysis and mobilization of liberated FFAs, resulting in reduced bacterial growth in adipocytes. Collectively, these results demonstrate that C. pneumoniae exploits host FABP4 to facilitate fat mobilization and intracellular replication in adipocytes. This work uncovers a novel strategy used by intracellular pathogens for acquiring energy via hijacking of the host lipid metabolism pathway.
Collapse
Affiliation(s)
- Nirwana Fitriani Walenna
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan; Department of Bacteriology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yusuke Kurihara
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Bin Chou
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Kazunari Ishii
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Toshinori Soejima
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Ryota Itoh
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Akinori Shimizu
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Takeshi Ichinohe
- Division of Viral Infection, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Kenji Hiromatsu
- Department of Microbiology & Immunology, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan.
| |
Collapse
|
47
|
Cai H, Wang T, Yang Z, Xu Z, Wang G, Wang HY, Zhu W, Chen K. Combined Virtual Screening and Substructure Search for Discovery of Novel FABP4 Inhibitors. J Chem Inf Model 2017; 57:2329-2335. [PMID: 28810126 DOI: 10.1021/acs.jcim.7b00364] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Fatty acid-binding protein 4 (FABP4, AFABP) is a potential drug target for diabetes and atherosclerosis. In this study, a series of novel FABP4 inhibitors were discovered through combining virtual screening and substructure search. Seventeen compounds exhibited FABP4 inhibitory activities with IC50 < 10 μM, among which 11 compounds showed high selectivity against FABP3. The best compound 36b displayed an IC50 value of 1.5 μM. Molecular docking and point mutation studies revealed that Gln95, Arg126, and Tyr128 play key roles for these compounds binding with FABP4. Interestingly, Gln95 seems to be essential for conformation stability of FABP4. The new scaffolds of these compounds and their interaction mechanisms binding with FABP4 should provide an important clue for the further development of novel FABP4 inhibitors.
Collapse
Affiliation(s)
- Haiyan Cai
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University, School of Medicine , Shanghai 200025, China.,Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | - Ting Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | - Zhuo Yang
- Chemical Biology Core Facility, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , Shanghai 200031, China
| | - Zhijian Xu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | - Guimin Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | - He-Yao Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | - Weiliang Zhu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| | - Kaixian Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road, Shanghai 201203, China
| |
Collapse
|
48
|
Rodríguez-Calvo R, Girona J, Alegret JM, Bosquet A, Ibarretxe D, Masana L. Role of the fatty acid-binding protein 4 in heart failure and cardiovascular disease. J Endocrinol 2017; 233:R173-R184. [PMID: 28420707 DOI: 10.1530/joe-17-0031] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 04/18/2017] [Indexed: 01/05/2023]
Abstract
Obesity and ectopic fat accumulation in non-adipose tissues are major contributors to heart failure (HF) and cardiovascular disease (CVD). Adipocytes act as endocrine organs by releasing a large number of bioactive molecules into the bloodstream, which participate in a communication network between white adipose tissue and other organs, including the heart. Among these molecules, fatty acid-binding protein 4 (FABP4) has recently been shown to increase cardiometabolic risk. Both clinical and experimental evidence have identified FABP4 as a relevant player in atherosclerosis and coronary artery disease, and it has been directly related to cardiac alterations such as left ventricular hypertrophy (LVH) and both systolic and diastolic cardiac dysfunction. The available interventional studies preclude the establishment of a direct causal role of this molecule in CVD and HF and propose FABP4 as a biomarker rather than as an aetiological factor. However, several experimental reports have suggested that FABP4 may act as a direct contributor to cardiac metabolism and physiopathology, and the pharmacological targeting of FABP4 may restore some of the metabolic alterations that are conducive to CVD and HF. Here, we review the current knowledge regarding FABP4 in the context of HF and CVD as well as the molecular basis by which this protein participates in the regulation of cardiac function.
Collapse
Affiliation(s)
- Ricardo Rodríguez-Calvo
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Josefa Girona
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Josep M Alegret
- Department of CardiologyCardiovascular Research Group, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Reus, Spain
| | - Alba Bosquet
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Daiana Ibarretxe
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| | - Lluís Masana
- Vascular Medicine and Metabolism UnitResearch Unit on Lipids and Atherosclerosis, 'Sant Joan' University Hospital, Universitat Rovira i Virgili, Institut de Investigació Sanitaria Pere Virgili (IISPV), Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Reus, Spain
| |
Collapse
|
49
|
Hertzel AV, Xu H, Downey M, Kvalheim N, Bernlohr DA. Fatty acid binding protein 4/aP2-dependent BLT1R expression and signaling. J Lipid Res 2017; 58:1354-1361. [PMID: 28546450 DOI: 10.1194/jlr.m074542] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 05/24/2017] [Indexed: 12/27/2022] Open
Abstract
Previous studies have shown that reduced levels of the adipocyte fatty acid binding protein (FABP)4 (AFABP/aP2), result in metabolic improvement including potentiated insulin sensitivity and attenuated atherosclerosis. Mechanistically, pharmacologic or genetic inhibition of FABP4 in macrophages upregulates UCP2, attenuates reactive oxygen species (ROS) production, polarizes cells toward the anti-inflammatory M2 state, and reduces leukotriene (LT) secretion. At the protein level, FABP4 stabilizes LTA4 toward chemical hydrolysis, thereby potentiating inflammatory LTC4 synthesis. Herein, we extend the FABP4-LT axis and demonstrate that genetic knockout of FABP4 reduces expression of the major macrophage LT receptor, LTB4 receptor 1 (BLT1R), via a ROS-dependent mechanism. Consistent with inflammation driving BLT1R expression, M1 polarized macrophages express increased levels of BLT1R relative to M2 polarized macrophages and treatment with proinflammatory lipopolysaccharide increased BLT1R mRNA and protein expression. In FABP4 knockout macrophages, silencing of UCP2, increased ROS levels and led to increased expression of BLT1R mRNA. Similarly, addition of exogenous H2O2 upregulated BLT1R expression, whereas the addition of a ROS scavenger, N-acetyl cysteine, decreased BLT1R levels. As compared with WT macrophages, LTB4-BLT1R-dependent JAK2-phosphorylation was reduced in FABP4 knockout macrophages. In summary, these results indicate that FABP4 regulates the expression of BLT1R and its downstream signaling via control of oxidative stress in macrophages.
Collapse
Affiliation(s)
- Ann V Hertzel
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | - Hongliang Xu
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | - Michael Downey
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | - Nicholas Kvalheim
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455.
| |
Collapse
|
50
|
Harjes U, Bridges E, Gharpure KM, Roxanis I, Sheldon H, Miranda F, Mangala LS, Pradeep S, Lopez-Berestein G, Ahmed A, Fielding B, Sood AK, Harris AL. Antiangiogenic and tumour inhibitory effects of downregulating tumour endothelial FABP4. Oncogene 2017; 36:912-921. [PMID: 27568980 PMCID: PMC5318662 DOI: 10.1038/onc.2016.256] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 05/15/2016] [Accepted: 06/01/2016] [Indexed: 12/31/2022]
Abstract
Fatty acid binding protein 4 (FABP4) is a fatty acid chaperone, which is induced during adipocyte differentiation. Previously we have shown that FABP4 in endothelial cells is induced by the NOTCH1 signalling pathway, the latter of which is involved in mechanisms of resistance to antiangiogenic tumour therapy. Here, we investigated the role of FABP4 in endothelial fatty acid metabolism and tumour angiogenesis. We analysed the effect of transient FABP4 knockdown in human umbilical vein endothelial cells on fatty acid metabolism, viability and angiogenesis. Through therapeutic delivery of siRNA targeting mouse FABP4, we investigated the effect of endothelial FABP4 knockdown on tumour growth and blood vessel formation. In vitro, siRNA-mediated FABP4 knockdown in endothelial cells led to a marked increase of endothelial fatty acid oxidation, an increase of reactive oxygen species and decreased angiogenesis. In vivo, we found that increased NOTCH1 signalling in tumour xenografts led to increased expression of endothelial FABP4 that decreased when NOTCH1 and VEGFA inhibitors were used in combination. Angiogenesis, growth and metastasis in ovarian tumour xenografts were markedly inhibited by therapeutic siRNA delivery targeting mouse endothelial FABP4. Therapeutic targeting of endothelial FABP4 by siRNA in vivo has antiangiogenic and antitumour effects with minimal toxicity and should be investigated further.
Collapse
MESH Headings
- Angiogenesis Inhibitors/metabolism
- Animals
- Apoptosis
- Biomarkers, Tumor/metabolism
- Cell Movement
- Cell Proliferation
- Cystadenocarcinoma, Serous/blood supply
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/pathology
- Cystadenocarcinoma, Serous/prevention & control
- Fatty Acid-Binding Proteins/antagonists & inhibitors
- Fatty Acid-Binding Proteins/genetics
- Fatty Acid-Binding Proteins/metabolism
- Female
- Follow-Up Studies
- Human Umbilical Vein Endothelial Cells/metabolism
- Human Umbilical Vein Endothelial Cells/pathology
- Humans
- Mice
- Mice, Nude
- Neoplasm Grading
- Neoplasm Invasiveness
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Ovarian Neoplasms/blood supply
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/prevention & control
- Prognosis
- Prospective Studies
- Receptor, Notch1/metabolism
- Signal Transduction
- Survival Rate
- Tumor Cells, Cultured
- Vascular Endothelial Growth Factor A/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- U Harjes
- Hypoxia and Growth Factor Group, WIMM, Department of Oncology, University of Oxford, Oxford, UK
| | - E Bridges
- Hypoxia and Growth Factor Group, WIMM, Department of Oncology, University of Oxford, Oxford, UK
| | - K M Gharpure
- Department of Gynecologic Oncology, University of Texas, Austin, TX, USA
| | - I Roxanis
- Department of Cellular Pathology, Oxford University Hospitals and NIHR Biomedical Research Centre Oxford, John Radcliffe Hospital, Oxford, UK
| | - H Sheldon
- Hypoxia and Growth Factor Group, WIMM, Department of Oncology, University of Oxford, Oxford, UK
| | - F Miranda
- Ovarian Cancer Cell Laboratory, WIMM, Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, UK
| | - L S Mangala
- Department of Gynecologic Oncology, University of Texas, Austin, TX, USA
- Center for RNA Interference and Non-Coding RNA, MD Anderson Cancer Center, Department of Gynecologic Oncology, University of Texas, Austin, TX, USA
| | - S Pradeep
- Department of Gynecologic Oncology, University of Texas, Austin, TX, USA
| | - G Lopez-Berestein
- Center for RNA Interference and Non-Coding RNA, MD Anderson Cancer Center, Department of Gynecologic Oncology, University of Texas, Austin, TX, USA
- Department of Cancer Biology, University of Texas, Austin, TX, USA
| | - A Ahmed
- Ovarian Cancer Cell Laboratory, WIMM, Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, UK
| | - B Fielding
- Department of Nutritional Sciences, University of Surrey, Surrey, UK
| | - A K Sood
- Department of Gynecologic Oncology, University of Texas, Austin, TX, USA
- Center for RNA Interference and Non-Coding RNA, MD Anderson Cancer Center, Department of Gynecologic Oncology, University of Texas, Austin, TX, USA
- Department of Cancer Biology, University of Texas, Austin, TX, USA
| | - A L Harris
- Hypoxia and Growth Factor Group, WIMM, Department of Oncology, University of Oxford, Oxford, UK
| |
Collapse
|