1
|
Dawid M, Pich K, Mlyczyńska E, Respekta-Długosz N, Wachowska D, Greggio A, Szkraba O, Kurowska P, Rak A. Adipokines in pregnancy. Adv Clin Chem 2024; 121:172-269. [PMID: 38797542 DOI: 10.1016/bs.acc.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Reproductive success consists of a sequential events chronology, starting with the ovum fertilization, implantation of the embryo, placentation, and cellular processes like proliferation, apoptosis, angiogenesis, endocrinology, or metabolic changes, which taken together finally conduct the birth of healthy offspring. Currently, many factors are known that affect the regulation and proper maintenance of pregnancy in humans, domestic animals, or rodents. Among the determinants of reproductive success should be distinguished: the maternal microenvironment, genes, and proteins as well as numerous pregnancy hormones that regulate the most important processes and ensure organism homeostasis. It is well known that white adipose tissue, as the largest endocrine gland in our body, participates in the synthesis and secretion of numerous hormones belonging to the adipokine family, which also may regulate the course of pregnancy. Unfortunately, overweight and obesity lead to the expansion of adipose tissue in the body, and its excess in both women and animals contributes to changes in the synthesis and release of adipokines, which in turn translates into dramatic changes during pregnancy, including those taking place in the organ that is crucial for the proper progress of pregnancy, i.e. the placenta. In this chapter, we are summarizing the current knowledge about levels of adipokines and their role in the placenta, taking into account the physiological and pathological conditions of pregnancy, e.g. gestational diabetes mellitus, preeclampsia, or intrauterine growth restriction in humans, domestic animals, and rodents.
Collapse
Affiliation(s)
- Monika Dawid
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Karolina Pich
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Ewa Mlyczyńska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Natalia Respekta-Długosz
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Dominka Wachowska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Krakow, Poland
| | - Aleksandra Greggio
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Oliwia Szkraba
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Patrycja Kurowska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Agnieszka Rak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland.
| |
Collapse
|
2
|
Orton S, Karkia R, Mustafov D, Gharanei S, Braoudaki M, Filipe A, Panfilov S, Saravi S, Khan N, Kyrou I, Karteris E, Chatterjee J, Randeva HS. In Silico and In Vitro Mapping of Receptor-Type Protein Tyrosine Phosphatase Receptor Type D in Health and Disease: Implications for Asprosin Signalling in Endometrial Cancer and Neuroblastoma. Cancers (Basel) 2024; 16:582. [PMID: 38339334 PMCID: PMC10854520 DOI: 10.3390/cancers16030582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Protein Tyrosine Phosphatase Receptor Type D (PTPRD) is involved in the regulation of cell growth, differentiation, and oncogenic transformation, as well as in brain development. PTPRD also mediates the effects of asprosin, which is a glucogenic hormone/adipokine derived following the cleavage of the C-terminal of fibrillin 1. Since the asprosin circulating levels are elevated in certain cancers, research is now focused on the potential role of this adipokine and its receptors in cancer. As such, in this study, we investigated the expression of PTPRD in endometrial cancer (EC) and the placenta, as well as in glioblastoma (GBM). METHODS An array of in silico tools, in vitro models, tissue microarrays (TMAs), and liquid biopsies were employed to determine the gene and protein expression of PTPRD in healthy tissues/organs and in patients with EC and GBM. RESULTS PTPRD exhibits high expression in the occipital lobe, parietal lobe, globus pallidus, ventral thalamus, and white matter, whereas in the human placenta, it is primarily localised around the tertiary villi. PTPRD is significantly upregulated at the mRNA and protein levels in patients with EC and GBM compared to healthy controls. In patients with EC, PTPRD is significantly downregulated with obesity, whilst it is also expressed in the peripheral leukocytes. The EC TMAs revealed abundant PTPRD expression in both low- and high-grade tumours. Asprosin treatment upregulated the expression of PTPRD only in syncytialised placental cells. CONCLUSIONS Our data indicate that PTPRD may have potential as a biomarker for malignancies such as EC and GBM, further implicating asprosin as a potential metabolic regulator in these cancers. Future studies are needed to explore the potential molecular mechanisms/signalling pathways that link PTPRD and asprosin in cancer.
Collapse
Affiliation(s)
- Sophie Orton
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK; (S.O.); (S.G.); (I.K.)
| | - Rebecca Karkia
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (R.K.); (D.M.); (A.F.); (S.P.); (S.S.); (N.K.); (E.K.)
| | - Denis Mustafov
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (R.K.); (D.M.); (A.F.); (S.P.); (S.S.); (N.K.); (E.K.)
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9JA, UK;
| | - Seley Gharanei
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK; (S.O.); (S.G.); (I.K.)
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
| | - Maria Braoudaki
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9JA, UK;
| | - Alice Filipe
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (R.K.); (D.M.); (A.F.); (S.P.); (S.S.); (N.K.); (E.K.)
| | - Suzana Panfilov
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (R.K.); (D.M.); (A.F.); (S.P.); (S.S.); (N.K.); (E.K.)
| | - Sayeh Saravi
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (R.K.); (D.M.); (A.F.); (S.P.); (S.S.); (N.K.); (E.K.)
| | - Nabeel Khan
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (R.K.); (D.M.); (A.F.); (S.P.); (S.S.); (N.K.); (E.K.)
| | - Ioannis Kyrou
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK; (S.O.); (S.G.); (I.K.)
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry CV1 5FB, UK
- College of Health, Psychology and Social Care, University of Derby, Derby DE22 1GB, UK
- Laboratory of Dietetics and Quality of Life, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
| | - Emmanouil Karteris
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (R.K.); (D.M.); (A.F.); (S.P.); (S.S.); (N.K.); (E.K.)
| | - Jayanta Chatterjee
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK; (R.K.); (D.M.); (A.F.); (S.P.); (S.S.); (N.K.); (E.K.)
- Academic Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust Hospital, Guildford GU2 7XX, UK
| | - Harpal S. Randeva
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK; (S.O.); (S.G.); (I.K.)
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry CV1 5FB, UK
| |
Collapse
|
3
|
Diao H, Li X, Xu Y, Xing X, Pang S. Asprosin, a novel glucogenic adipokine implicated in type 2 diabetes mellitus. J Diabetes Complications 2023; 37:108614. [PMID: 37769508 DOI: 10.1016/j.jdiacomp.2023.108614] [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: 05/05/2023] [Revised: 09/12/2023] [Accepted: 09/17/2023] [Indexed: 10/03/2023]
Abstract
Asprosin, encoded by penultimate two exons (exon 65 and exon 66) of the gene Fibrillin 1 (FBN1), has been recently discovered to be a novel hormone secreted by white adipose tissues during fasting. The glucose metabolism disorders are often accompanied by increased asprosin level. Previous research suggests that asprosin may contribute to the development of diabetes by regulating glucose homeostasis, appetite, insulin secretion, and insulin sensitivity. In this review, we summarize the recent findings from studies on asprosin and its association with Type 2 diabetes mellitus, and discusses its mechanisms from various aspects, so as to provide clinical diagnosis and treatment ideas for T2DM.
Collapse
Affiliation(s)
- Hongcui Diao
- Department of Endocrinology, Jinan Central Hospital, Shandong University, Jinan, China
| | - Xue Li
- Department of Infectious Diseases, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yeqiu Xu
- Department of Eye, Oral & Plastic Surgery, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiuli Xing
- Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shuguang Pang
- Department of Endocrinology, Jinan Central Hospital, Shandong University, Jinan, China; Department of Endocrinology, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
4
|
Boz İB, Aytürk Salt S, Salt Ö, Sayın NC, Dibirdik İ. Association Between Plasma Asprosin Levels and Gestational Diabetes Mellitus. Diabetes Metab Syndr Obes 2023; 16:2515-2521. [PMID: 37641645 PMCID: PMC10460609 DOI: 10.2147/dmso.s424651] [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] [Received: 06/06/2023] [Accepted: 08/10/2023] [Indexed: 08/31/2023] Open
Abstract
Purpose This study sought to investigate whether asprosin can be used in the diagnosis of GDM or for diagnostic purposes in high-risk pregnancies, along with a review of other parameters that may be associated with serum asprosin levels. Patients and Methods The study investigated the association between gestational diabetes mellitus (GDM) and asprosin levels. A total of 93 participants; 30 patients with GDM, 33 healthy pregnant women with normal glucose tolerance (NGT), and 30 healthy non-diabetic women (control group) at the Endocrinology and Metabolic Diseases outpatient clinic of a tertiary care university hospital were enrolled in the study. Patients with GDM and NGT were examined in terms of GDM between the 24th and 28th week of pregnancy (2nd trimester). Patient data were collected during routine examinations, and asprosin levels were measured using the ELISA method. All participants underwent testing for measurements of serum hemoglobin, insulin, C-peptide, fasting plasma glucose, and glycated hemoglobin (HbA1c) levels following a fasting period of at least eight hours. Results Asprosin levels were higher in pregnant women with NGT and with GDM versus controls (Control-NGT asprosin, p = 0.001; Control-GDM asprosin, p = 0.001). Pregnant women with GDM had higher asprosin levels than those with NGT (p = 0.001). In detecting GDM in pregnant women, an asprosin cutoff value of >31.709 ng/mL yielded a sensitivity of 93.3%, specificity of 90.9%, positive predictive value of 90.3%, and negative predictive value of 93.75% (p < 0.001). Conclusion Serum asprosin levels can potentially be used as a marker in the diagnosis of GDM.
Collapse
Affiliation(s)
| | - Semra Aytürk Salt
- Department of Endocrinology and Metabolic Diseases, Kayseri City Hospital, Kayseri, Turkey
| | - Ömer Salt
- Department of Emergency Medicine, Kayseri City Hospital, Kayseri, Turkey
| | - Niyazi Cenk Sayın
- Department of Department of Obstetrics and Gynecology, Trakya University, Edirne, Turkey
| | - İlker Dibirdik
- Department of Clinical Biochemistry, Trakya University, Edirne, Turkey
| |
Collapse
|
5
|
Yavuz A, Aydin MA, Ugur K, Aydin S, Senol A, Baykus Y, Deniz R, Sahin İ, Yalcin MH, Gencer BT, Deniz YK, Ustebay S, Karagoz ZK, Emre E, Aydin S. Betatrophin, elabela, asprosin, glucagon and subfatin peptides in breast tissue, blood and milk in gestational diabetes. Biotech Histochem 2023; 98:243-254. [PMID: 36825397 DOI: 10.1080/10520295.2023.2176546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
We investigated the presence of asprosin (ASP), betatrophin, elabela (ELA), glucagon and subfatin (SUB) in the milk of mothers with gestational diabetes mellitus (GDM) and compared their levels with blood levels. We also investigated whether these peptides are synthesized by the breast. We investigated 12 volunteer mothers with GDM and 14 pregnant non-GDM control mothers. The peptides were measured using ELISA and their tissue localization was determined using immunohistochemistry. Breast milk contains ASP, betatrophin, ELA, glucagon and SUB. The amount of the peptides ranged from highest to the lowest in colostrum, transitional milk and mature milk. The amount of peptides in the milk was greater than for blood. The peptides, except for ELA, were increased in milk and blood by GDM. Betatrophin and ELA are synthesized in the connective tissue of the breast. ASP, glucagon and SUB are synthesized in the alveolar tissue of the breast. These peptides in breast milk may contribute to the development of the gastrointestinal tract of newborns and infants.
Collapse
Affiliation(s)
- Adem Yavuz
- Department of Obstetrics and Gynecology, Nigde Omer Halis Demir Research and Education Hospital, Nigde, Turkiye
| | - Mustafa Ata Aydin
- Medical Student, School of Medicine, Gazi University, Ankara, Turkiye
| | - Kader Ugur
- Department of Internal Medicine (Endocrinology and Metabolism Diseases), School of Medicine, Firat University, Elazig, Turkiye
| | - Suna Aydin
- Department of Cardiovascular Surgery, Fethi Sekin City Hospital, Elazig, Turkiye
- Department of Anatomy, School of Medicine, Firat University, Elazig, Turkiye
- Department of Histology and Embryology, School of Veterinary Medicine, Firat University, Elazig, Turkiye
| | - Arzu Senol
- Department of Enfection Disease, Fethi Sekin City Hospital, Elazig, Turkiye
| | - Yakup Baykus
- Department of Obstetrics and Gynecology, Bandirma 17 Eylul Univerity, Balikesir, Turkiye
| | - Rulin Deniz
- Department of Obstetrics and Gynecology, Bandirma 17 Eylul Univerity, Balikesir, Turkiye
| | - İbrahim Sahin
- Department of Medical Biochemistry and Clinical Biochemistry, (Firat Hormones Research Group), Medical School, Firat University, Elazig, Turkiye
- Department of Medical Biology, School of Medicine, Erzincan Binali Yildirim University, Erzincan, Turkiye
| | - Mehmet Hanifi Yalcin
- Department of Histology and Embryology, School of Veterinary Medicine, Firat University, Elazig, Turkiye
| | - Berrin Tarakci Gencer
- Department of Histology and Embryology, School of Veterinary Medicine, Firat University, Elazig, Turkiye
| | - Yaprak Kandemir Deniz
- Department of Obstetrics and Gynecology, Antalya Medicalpark Hospital Complex, Antalya, Turkiye
| | - Sefer Ustebay
- Department of Pediatrics, Bandirma 17 Eylul Univerity, Balikesir, Turkiye
| | - Zuhal Karaca Karagoz
- Department of Internal Medicine (Endocrinology and Metabolism Diseases), Fethi Sekin City Hospital, Elazig, Turkiye
| | - Elif Emre
- Department of Anatomy, School of Medicine, Firat University, Elazig, Turkiye
| | - Suleyman Aydin
- Department of Medical Biochemistry and Clinical Biochemistry, (Firat Hormones Research Group), Medical School, Firat University, Elazig, Turkiye
| |
Collapse
|
6
|
Farrag M, Ait Eldjoudi D, González-Rodríguez M, Cordero-Barreal A, Ruiz-Fernández C, Capuozzo M, González-Gay MA, Mera A, Lago F, Soffar A, Essawy A, Pino J, Farrag Y, Gualillo O. Asprosin in health and disease, a new glucose sensor with central and peripheral metabolic effects. Front Endocrinol (Lausanne) 2023; 13:1101091. [PMID: 36686442 PMCID: PMC9849689 DOI: 10.3389/fendo.2022.1101091] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023] Open
Abstract
Adipose tissue malfunction leads to altered adipokine secretion which might consequently contribute to an array of metabolic diseases spectrum including obesity, diabetes mellitus, and cardiovascular disorders. Asprosin is a novel diabetogenic adipokine classified as a caudamin hormone protein. This adipokine is released from white adipose tissue during fasting and elicits glucogenic and orexigenic effects. Although white adipose tissue is the dominant source for this multitask adipokine, other tissues also may produce asprosin such as salivary glands, pancreatic B-cells, and cartilage. Significantly, plasma asprosin levels link to glucose metabolism, lipid profile, insulin resistance (IR), and β-cell function. Indeed, asprosin exhibits a potent role in the metabolic process, induces hepatic glucose production, and influences appetite behavior. Clinical and preclinical research showed dysregulated levels of circulating asprosin in several metabolic diseases including obesity, type 2 diabetes mellitus (T2DM), polycystic ovarian syndrome (PCOS), non-alcoholic fatty liver (NAFLD), and several types of cancer. This review provides a comprehensive overview of the asprosin role in the etiology and pathophysiological manifestations of these conditions. Asprosin could be a promising candidate for both novel pharmacological treatment strategies and diagnostic tools, although developing a better understanding of its function and signaling pathways is still needed.
Collapse
Affiliation(s)
- Mariam Farrag
- SERGAS (Servizo Galego de Saude), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
- Euro-Mediterranean Master in neuroscience and Biotechnology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Djedjiga Ait Eldjoudi
- SERGAS (Servizo Galego de Saude), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - María González-Rodríguez
- SERGAS (Servizo Galego de Saude), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
- International PhD School of the University of Santiago de Compostela (EDIUS), Doctoral Program in Drug Research and Development, Santiago de Compostela, Spain
| | - Alfonso Cordero-Barreal
- SERGAS (Servizo Galego de Saude), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Clara Ruiz-Fernández
- SERGAS (Servizo Galego de Saude), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
- International PhD School of the University of Santiago de Compostela (EDIUS), Doctoral Program in Medicine Clinical Research, Santiago de Compostela, Spain
| | - Maurizio Capuozzo
- National Health Service, Local Health Authority ASL 3 Napoli Sud, Department of Pharmacy, Naples, Italy
| | - Miguel Angel González-Gay
- Hospital Universitario Marqués de Valdecilla, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, IDIVAL, University of Cantabria, Santander, Cantabria, Spain
| | - Antonio Mera
- SERGAS, Santiago University Clinical Hospital, Division of Rheumatology, Santiago de Compostela, Spain
| | - Francisca Lago
- SERGAS (Servizo Galego de Saude), IDIS (Instituto de Investigación Sanitaria de Santiago), Molecular and Cellular Cardiology Lab, Research Laboratory 7, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Ahmed Soffar
- Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Amina Essawy
- Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Jesus Pino
- SERGAS (Servizo Galego de Saude), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Yousof Farrag
- SERGAS (Servizo Galego de Saude), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| |
Collapse
|
7
|
Summers KM, Bush SJ, Davis MR, Hume DA, Keshvari S, West JA. Fibrillin-1 and asprosin, novel players in metabolic syndrome. Mol Genet Metab 2023; 138:106979. [PMID: 36630758 DOI: 10.1016/j.ymgme.2022.106979] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Fibrillin-1 is a major component of the extracellular microfibrils, where it interacts with other extracellular matrix proteins to provide elasticity to connective tissues, and regulates the bioavailability of TGFβ family members. A peptide consisting of the C-terminal 140 amino acids of fibrillin-1 has recently been identified as a glucogenic hormone, secreted from adipose tissue during fasting and targeting the liver to release glucose. This fragment, called asprosin, also signals in the hypothalamus to stimulate appetite. Asprosin levels are correlated with many of the pathologies indicative of metabolic syndrome, including insulin resistance and obesity. Previous studies and reviews have addressed the therapeutic potential of asprosin as a target in obesity, diabetes and related conditions without considering mechanisms underlying the relationship between generation of asprosin and expression of the much larger fibrillin-1 protein. Profibrillin-1 undergoes obligatory cleavage at the cell surface as part of its assembly into microfibrils, producing the asprosin peptide as well as mature fibrillin-1. Patterns of FBN1 mRNA expression are inconsistent with the necessity for regulated release of asprosin. The asprosin peptide may be protected from degradation in adipose tissue. We present evidence for an alternative possibility, that asprosin mRNA is generated independently from an internal promoter within the 3' end of the FBN1 gene, which would allow for regulation independent of fibrillin-synthesis and is more economical of cellular resources. The discovery of asprosin opened exciting possibilities for treatment of metabolic syndrome related conditions, but there is much to be understood before such therapies could be introduced into the clinic.
Collapse
Affiliation(s)
- Kim M Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, Queensland 4102, Australia.
| | - Stephen J Bush
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford OX3 9DS, United Kingdom.
| | - Margaret R Davis
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, United Kingdom
| | - David A Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, Queensland 4102, Australia.
| | - Sahar Keshvari
- Mater Research Institute-University of Queensland, Translational Research Institute, 37 Kent St, Woolloongabba, Queensland 4102, Australia.
| | - Jennifer A West
- Faculty of Medicine, The University of Queensland, Mayne Medical Building, 288 Herston Road, Herston, Queensland 4006, Australia.
| |
Collapse
|
8
|
Differential Regulation of Genes by the Glucogenic Hormone Asprosin in Ovarian Cancer. J Clin Med 2022; 11:jcm11195942. [PMID: 36233808 PMCID: PMC9573256 DOI: 10.3390/jcm11195942] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 11/07/2022] Open
Abstract
Background: Ovarian cancer (OvCa) is one of the most lethal forms of gynaecological malignancy. Altered energy metabolism and increased aerobic glycolysis in OvCa are hallmarks that demand attention. The glucogenic hormone asprosin is often dysregulated in metabolic disorders such as insulin resistance, diabetes (type 2 and gestational), and preeclampsia. Despite association with metabolic disorders, its role in energy metabolism within the tumour microenvironment is yet to be explored. Here, we study the role of asprosin in OvCa using transcriptomics and expand on functional studies with clinical samples. Methods: RNA sequencing, functional gene enrichment analysis, Western blotting and ImageStream. Results: Following treatment with 100 nM of asprosin, the serous OvCa cell line, SKOV-3, displayed 160 and 173 gene regulatory changes, at 4 and 12 h respectively, when compared with control samples (p < 0.05 and Log2FC > 1). In addition to energy metabolism and glucose-related pathways, asprosin was shown to alter pathways associated with cell communication, TGF-β signalling, and cell proliferation. Moreover, asprosin was shown to induce phosphorylation of ERK1/2 in the same in vitro model. Using liquid biopsies, we also report for novel expression of asprosin’s predicted receptors OR4M1 and TLR4 in cancer-associated circulating cells; with significant reduction seen between pre-chemotherapy and end of first line chemotherapy, in addition to patients under maintenance with bevacizumab +/− olaparib for OR4M1. Conclusions: In relation to OvCa, asprosin appears to regulate numerous signalling pathways in-vitro. The prognostic potential of OR4M1 in liquid biopsies should also be explored further.
Collapse
|
9
|
Liu L, Liu Y, Huang M, Zhang M, Zhu C, Chen X, Bennett S, Xu J, Zou J. The Effects of Asprosin on Exercise-Intervention in Metabolic Diseases. Front Physiol 2022; 13:907358. [PMID: 35899030 PMCID: PMC9311488 DOI: 10.3389/fphys.2022.907358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Fibrillin is the major constituent of extracellular microfibrils, which are distributed throughout connective tissues. Asprosin is derived from the C-terminal region of the FBN1 gene, which encodes profibrillin that undergoes cleavage by furin protein. In response to fasting with low dietary glucose, asprosin is released as a secreted factor from white adipose tissue, and is transported to the liver for the mediation of glucose release into the blood circulation. Through binding to OLFR734, an olfactory G-protein-coupled receptor in liver cells, asprosin induces a glucogenic effect to regulate glucose homeostasis. Bioinformatics analyses revealed that the FBN1 gene is abundantly expressed in human skeletal muscle-derived mesoangioblasts, osteoblast-like cells, and mesenchymal stem cells, indicating that the musculoskeletal system might play a role in the regulation of asprosin expression. Interestingly, recent studies suggest that asprosin is regulated by exercise. This timely review discusses the role of asprosin in metabolism, its receptor signalling, as well as the exercise regulation of asprosin. Collectively, asprosin may have a vital regulatory effect on the improvement of metabolic disorders such as diabetes mellitus and obesity via exercise.
Collapse
Affiliation(s)
- Lifei Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation, The People’s Hospital of Liaoning Province, Shenyang, China
| | - Yuhao Liu
- Department of Orthopaedic, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Mei Huang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Miao Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Chenyu Zhu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Xi Chen
- School of Sports Science, Wenzhou Medical University, Wenzhou, China
| | - Samuel Bennett
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
- *Correspondence: Jun Zou, ; Jiake Xu,
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- *Correspondence: Jun Zou, ; Jiake Xu,
| |
Collapse
|
10
|
Mishra I, Xie WR, Bournat JC, He Y, Wang C, Silva ES, Liu H, Ku Z, Chen Y, Erokwu BO, Jia P, Zhao Z, An Z, Flask CA, He Y, Xu Y, Chopra AR. Protein tyrosine phosphatase receptor δ serves as the orexigenic asprosin receptor. Cell Metab 2022; 34:549-563.e8. [PMID: 35298903 PMCID: PMC8986618 DOI: 10.1016/j.cmet.2022.02.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/25/2021] [Accepted: 02/22/2022] [Indexed: 12/20/2022]
Abstract
Asprosin is a fasting-induced glucogenic and centrally acting orexigenic hormone. The olfactory receptor Olfr734 is known to be the hepatic receptor for asprosin that mediates its effects on glucose production, but the receptor for asprosin's orexigenic function has been unclear. Here, we have identified protein tyrosine phosphatase receptor δ (Ptprd) as the orexigenic receptor for asprosin. Asprosin functions as a high-affinity Ptprd ligand in hypothalamic AgRP neurons, regulating the activity of this circuit in a cell-autonomous manner. Genetic ablation of Ptprd results in a strong loss of appetite, leanness, and an inability to respond to the orexigenic effects of asprosin. Ablation of Ptprd specifically in AgRP neurons causes resistance to diet-induced obesity. Introduction of the soluble Ptprd ligand-binding domain in the circulation of mice suppresses appetite and blood glucose levels by sequestering plasma asprosin. Identification of Ptprd as the orexigenic asprosin receptor creates a new avenue for the development of anti-obesity therapeutics.
Collapse
Affiliation(s)
- Ila Mishra
- Harrington Discovery Institute, Cleveland, OH, USA
| | - Wei Rose Xie
- Harrington Discovery Institute, Cleveland, OH, USA
| | - Juan C Bournat
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yang He
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Chunmei Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | - Hailan Liu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Zhiqiang Ku
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yinghua Chen
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, USA
| | - Bernadette O Erokwu
- Departments of Radiology, Biomedical Engineering, and Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhiqiang An
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chris A Flask
- Departments of Radiology, Biomedical Engineering, and Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Yanlin He
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Yong Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Atul R Chopra
- Harrington Discovery Institute, Cleveland, OH, USA; Department of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
11
|
Hoffmann T, Morcos YAT, Janoschek R, Turnwald EM, Gerken A, Müller A, Sengle G, Dötsch J, Appel S, Hucklenbruch-Rother E. Correlation of metabolic characteristics with maternal, fetal and placental asprosin in human pregnancy. Endocr Connect 2022; 11:e220069. [PMID: 35148275 PMCID: PMC8942321 DOI: 10.1530/ec-22-0069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/11/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Asprosin is a recently discovered hormone associated with obesity and diabetes mellitus. Little is known about asprosin's role during pregnancy, but a contribution of asprosin to pregnancy complications resulting from maternal obesity and gestational diabetes mellitus (GDM) is conceivable. We assessed the potential effects of obesity, GDM and other clinical parameters on maternal and fetal umbilical plasma asprosin concentrations and placental asprosin expression. DESIGN The Cologne-Placenta Cohort Study comprises 247 female patients, from whom blood and placentas were collected at the University Hospital Cologne. METHODS We studied the maternal and fetal umbilical plasma and placentas of pregnant women with an elective, primary section. Sandwich ELISA measurements of maternal and fetal umbilical plasma and immunohistochemical stainings of placental tissue were performed to determine the asprosin levels. Also, the relation between asprosin levels and clinical blood parameters was studied. RESULTS There was a strong correlation between the maternal and fetal plasma asprosin levels and both increased with GDM in normal-weight and obese women. Asprosin immunoreactivity was measured in cultivated placental cells and placental tissue. BMI and GDM were not but pre-pregnancy exercise and smoking were correlated with maternal and/or fetal asprosin levels. Placental asprosin levels were associated with maternal but not with fetal plasma asprosin levels and with BMI but not with GDM. Placental asprosin was related to maternal insulin levels and increased upon insulin treatment in GDM patients. CONCLUSIONS Asprosin could potentially act as a biomarker and contribute to the clinical manifestation of pregnancy complications associated with maternal obesity.
Collapse
Affiliation(s)
- Thorben Hoffmann
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Yousef Ashraf Tawfik Morcos
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ruth Janoschek
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Eva-Maria Turnwald
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Antje Gerken
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Annette Müller
- Center for Pediatric Pathology at the University Hospital Cologne, Cologne, Germany
| | - Gerhard Sengle
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Cologne Center for Musculoskeletal Biomechanics (CCMB), Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Sarah Appel
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Eva Hucklenbruch-Rother
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Correspondence should be addressed to E Hucklenbruch-Rother:
| |
Collapse
|
12
|
Behrasi F, Karajibani M, Khayat S, Fanaei H, Montazerifar F. Association of maternal and umbilical cord blood asprosin with excessive gestational weight gain. NUTR BULL 2022; 47:50-56. [DOI: 10.1111/nbu.12542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Fatemeh Behrasi
- Department of Nutrition School of Medicine Zahedan University of Medical Sciences Zahedan Iran
| | - Mansour Karajibani
- Department of Nutrition Health Promotion Research Center School of Medicine Zahedan University of Medical Sciences Zahedan Iran
| | - Samira Khayat
- Department of Midwifery, Pregnancy Health Research Center School of Nursing and Midwifery Zahedan University of Medical Sciences Zahedan Iran
| | - Hamed Fanaei
- Department of Physiology Pregnancy Health Research Center School of Medicine Zahedan University of Medical Sciences Zahedan Iran
| | - Farzaneh Montazerifar
- Department of Nutrition Pregnancy Health Research Center School of Medicine Zahedan University of Medical Sciences Zahedan Iran
| |
Collapse
|
13
|
Morcos YAT, Lütke S, Tenbieg A, Hanisch FG, Pryymachuk G, Piekarek N, Hoffmann T, Keller T, Janoschek R, Niehoff A, Zaucke F, Dötsch J, Hucklenbruch-Rother E, Sengle G. Sensitive asprosin detection in clinical samples reveals serum/saliva correlation and indicates cartilage as source for serum asprosin. Sci Rep 2022; 12:1340. [PMID: 35079041 PMCID: PMC8789892 DOI: 10.1038/s41598-022-05060-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023] Open
Abstract
The C-terminal pro-fibrillin-1 propeptide asprosin is described as white adipose tissue derived hormone that stimulates rapid hepatic glucose release and activates hunger-promoting hypothalamic neurons. Numerous studies proposed correlations of asprosin levels with clinical parameters. However, the enormous variability of reported serum and plasma asprosin levels illustrates the need for sensitive and reliable detection methods in clinical samples. Here we report on newly developed biochemical methods for asprosin concentration and detection in several body fluids including serum, plasma, saliva, breast milk, and urine. Since we found that glycosylation impacts human asprosin detection we analyzed its glycosylation profile. Employing a new sandwich ELISA revealed that serum and saliva asprosin correlate strongly, depend on biological sex, and feeding status. To investigate the contribution of connective tissue-derived asprosin to serum levels we screened two cohorts with described cartilage turnover. Serum asprosin correlated with COMP, a marker for cartilage degradation upon running exercise and after total hip replacement surgery. This together with our finding that asprosin is produced by primary human chondrocytes and expressed in human cartilage suggests a contribution of cartilage to serum asprosin. Furthermore, we determined asprosin levels in breast milk, and urine, for the first time, and propose saliva asprosin as an accessible clinical marker for future studies.
Collapse
Affiliation(s)
- Yousef A T Morcos
- Center for Biochemistry, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Joseph-Stelzmann-Street 52, 50931, Cologne, Germany
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Steffen Lütke
- Center for Biochemistry, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Joseph-Stelzmann-Street 52, 50931, Cologne, Germany
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Antje Tenbieg
- Center for Biochemistry, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Joseph-Stelzmann-Street 52, 50931, Cologne, Germany
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Franz-Georg Hanisch
- Center for Biochemistry, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Joseph-Stelzmann-Street 52, 50931, Cologne, Germany
| | - Galyna Pryymachuk
- Department of Anatomy I, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nadin Piekarek
- Department of Anatomy I, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thorben Hoffmann
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Titus Keller
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ruth Janoschek
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Anja Niehoff
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, Cologne, Germany
- Cologne Center for Musculoskeletal Biomechanics (CCMB), Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Department of Orthopaedics (Friedrichsheim), University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Eva Hucklenbruch-Rother
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gerhard Sengle
- Center for Biochemistry, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Joseph-Stelzmann-Street 52, 50931, Cologne, Germany.
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Cologne Center for Musculoskeletal Biomechanics (CCMB), Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
14
|
Yang Z, Jiang J, Chen M, Huang J, Liu J, Wei X, Jia R, Song L, Sun B, Luo X, Song Q, Han Z. Sex-Specific Effects of Maternal and Post-Weaning High-Fat Diet on Adipose Tissue Remodeling and Asprosin Expression in Mice Offspring. Mol Nutr Food Res 2021; 66:e2100470. [PMID: 34933410 DOI: 10.1002/mnfr.202100470] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/15/2021] [Indexed: 11/07/2022]
Abstract
SCOPE Perinatal high-fat diet (HFD) increases risk of metabolic disorders in offspring. Adipose tissue remodeling is associated with metabolic syndrome. The current study characterizes the profile of maternal HFD-induced changes in adipose tissue remodeling and adipokines expression in mice offspring. METHODS AND RESULTS Female C57BL/6 mice are fed with CHOW or HFD for 2 weeks before mating, throughout gestation and lactation. At weaning, pups are randomly fed with CHOW or HFD, resulting in eight groups according to sex and maternal diet: Male CHOW-CHOW (MCC), Male CHOW-HFD (MCH), Male HFD-CHOW (MHC), Male HFD-HFD (MHH), Female CHOW-CHOW (FCC), Female CHOW-HFD (FCH), Female HFD-CHOW (FHC), and Female HFD-HFD (FHH). Increased body weight, impaired glucose tolerance, increased adipose tissue mass and hypertrophy, and decreased circulating asprosin level are only observed in male offspring exposure to maternal HFD. Serum asprosin level negatively correlates with fasting blood glucose, serum cholesterol (CHO), and high-density lipoprotein (HDL) levels, while positively correlates with serum low-density lipoprotein (LDL) and glutamate-oxaloacetate transaminase (GOT) levels in male offspring. A combination of genetic and biochemical analyses of adipokines shows the depot- and sex-specific changes in response to maternal and/or post-weaning HFD. CONCLUSION This study's results reveal the differential metabolic changes in response to maternal and/or post-weaning HFD in male and female offspring. The effect of maternal HFD on metabolic phonotype is more obvious in male offspring, supporting the notion that males are more susceptible to HFD-induced metabolic disorders.
Collapse
Affiliation(s)
- Zhao Yang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jianan Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Miao Chen
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jiaqi Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Jing Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Xiaojing Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Ru Jia
- Department of Prosthodontics, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lin Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Bo Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Xiao Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.,Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China
| | - Qing Song
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhen Han
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
15
|
Moradi N, Fouani FZ, Vatannejad A, Bakhti Arani A, Shahrzad S, Fadaei R. Serum levels of Asprosin in patients diagnosed with coronary artery disease (CAD): a case-control study. Lipids Health Dis 2021; 20:88. [PMID: 34419063 PMCID: PMC8380384 DOI: 10.1186/s12944-021-01514-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/01/2021] [Indexed: 01/02/2023] Open
Abstract
Background Coronary artery disease (CAD) is considered as a multi-faceted chronic inflammatory disease involving reduced blood supply to the myocardium as a result of accumulating lipids in the atrial walls. Visceral adiposity with disrupted release of adipokines play a key role in its pathogenesis. Asprosin is a newly identified fasting-induced glucogenic adipokine that has been related with metabolic disorders such as type II diabetes mellitus and polycystic ovary syndrome. The preset study sought to assess circulating asprosin in context of CAD. Methods In this study, serum levels of asprosin were determined in 88 CAD patients and 88 non-CAD healthy controls. Serum IL-6, TNF-α, asprosin and adiponectin were assessed using ELISA kits. Results: Serum asprosin was found to be higher in CAD patients when compared to non-CAD subjects (7.84 ± 2.08 versus 5.02 ± 1.29 μg/mL, p < 0.001). Similarly, serum TNF-α, and IL-6 elevated in CAD group significantly (p < 0.001). However, circulating adiponectin diminished in CAD group when compared with non-CAD subjects (p < 0.001). Moreover, serum asprosin levels directly correlated with BMI, FBG, HOMA-IR, TG and TC. Logistic regression analyses showed that asprosin levels were associated with increased risk of developing CAD (odds ratio: 3.01, 95% CI: 2.16, 4.20 and p < 0.001), after adjusting for potential confounders (age, sex and BMI). Conclusions The present study findings suggested a possible relation of serum asprosin with the pathogenesis of CAD, in particular through insulin resistance and dyslipidemia.
Collapse
Affiliation(s)
- Nariman Moradi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Fatima Zahraa Fouani
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Vatannejad
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Abbas Bakhti Arani
- Department of Cardiology, Dr Shariatee training and research Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Soraya Shahrzad
- Department of Cardiology, Dr Shariatee training and research Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Reza Fadaei
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
16
|
Shabir K, Brown JE, Afzal I, Gharanei S, Weickert MO, Barber TM, Kyrou I, Randeva HS. Asprosin, a novel pleiotropic adipokine implicated in fasting and obesity-related cardio-metabolic disease: Comprehensive review of preclinical and clinical evidence. Cytokine Growth Factor Rev 2021; 60:120-132. [PMID: 34108103 DOI: 10.1016/j.cytogfr.2021.05.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/09/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022]
Abstract
White adipose tissue is a dynamic endocrine organ that releases an array of adipokines, which play a key role in regulating metabolic homeostasis and multiple other physiological processes. An altered adipokine secretion profile from adipose tissue depots frequently characterizes obesity and related cardio-metabolic diseases. Asprosin is a recently discovered adipokine that is released in response to fasting. Following secretion, asprosin acts - via an olfactory G-protein coupled receptor and potentially via other unknown receptor(s) - on hepatocytes and agouti-related peptide-expressing neurons in the central nervous system to stimulate glucose secretion and promote appetite, respectively. A growing body of both in vitro and in vivo studies have shown asprosin to exert a number of effects on different metabolic tissues. Indeed, asprosin can attenuate insulin signalling and promote insulin resistance in skeletal muscle by increasing inflammation and endoplasmic reticulum stress. Interestingly, asprosin may also play a protective role in cardiomyocytes that are exposed to hypoxic conditions. Moreover, clinical studies have reported elevated circulating asprosin levels in obesity, type 2 diabetes and other obesity-related cardio-metabolic diseases, with significant associations to clinically relevant parameters. Understanding the spectrum of the effects of this novel adipokine is essential in order to determine its physiologic role and its significance as a potential therapeutic target and/or a biomarker of cardio-metabolic disease. The present review offers a comprehensive overview of the published literature on asprosin, including both clinical and preclinical studies, focusing on its role in metabolism and cardio-metabolic disease.
Collapse
Affiliation(s)
- Kiran Shabir
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, United Kingdom
| | - James E Brown
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, United Kingdom; School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, United Kingdom
| | - Islam Afzal
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, United Kingdom
| | - Seley Gharanei
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Martin O Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom; Centre of Applied Biological & Exercise Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry, CV1 5FB, United Kingdom
| | - Thomas M Barber
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, United Kingdom; Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom; Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, CV1 5FB, United Kingdom.
| | - Harpal S Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, United Kingdom; Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom.
| |
Collapse
|
17
|
Mishra I, Duerrschmid C, Ku Z, He Y, Xie W, Silva ES, Hoffman J, Xin W, Zhang N, Xu Y, An Z, Chopra AR. Asprosin-neutralizing antibodies as a treatment for metabolic syndrome. eLife 2021; 10:63784. [PMID: 33904407 PMCID: PMC8102062 DOI: 10.7554/elife.63784] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Background Recently, we discovered a new glucogenic and centrally acting orexigenic hormone - asprosin. Asprosin is elevated in metabolic syndrome (MS) patients, and its genetic loss results in reduced appetite, leanness, and blood glucose burden, leading to protection from MS. Methods We generated three independent monoclonal antibodies (mAbs) that recognize unique asprosin epitopes and investigated their preclinical efficacy and tolerability in the treatment of MS. Results Anti-asprosin mAbs from three distinct species lowered appetite and body weight, and reduced blood glucose in a dose-dependent and epitope-agnostic fashion in three independent MS mouse models, with an IC50 of ~1.5 mg/kg. The mAbs displayed a half-life of over 3days in vivo, with equilibrium dissociation-constants in picomolar to low nanomolar range. Conclusions We demonstrate that anti-asprosin mAbs are dual-effect pharmacologic therapy that targets two key pillars of MS - over-nutrition and hyperglycemia. This evidence paves the way for further development towards an investigational new drug application and subsequent human trials for treatment of MS, a defining physical ailment of our time. Funding DK118290 and DK125403 (R01; National Institute of Diabetes and Digestive and Kidney Diseases), DK102529 (K08; National Institute of Diabetes and Digestive and Kidney Diseases), Caroline Wiess Law Scholarship (Baylor College of Medicine, Harrington Investigatorship Harrington Discovery Institute at University Hospitals, Cleveland); Chao Physician Scientist Award (Baylor College of Medicine); RP150551 and RP190561 (Cancer Prevention and Research Institute of Texas [CPRIT]).
Collapse
Affiliation(s)
- Ila Mishra
- Harrington Discovery Institute, University Hospitals, Cleveland, United States
| | - Clemens Duerrschmid
- Harrington Discovery Institute, University Hospitals, Cleveland, United States
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, United States
| | - Yang He
- Baylor College of Medicine, Houston, United States
| | - Wei Xie
- Harrington Discovery Institute, University Hospitals, Cleveland, United States
| | | | - Jennifer Hoffman
- Harrington Discovery Institute, University Hospitals, Cleveland, United States
| | - Wei Xin
- Department of Pathology, Case Western Reserve University, Cleveland, United States
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, United States
| | - Yong Xu
- Baylor College of Medicine, Houston, United States
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, United States
| | - Atul R Chopra
- Harrington Discovery Institute, University Hospitals, Cleveland, United States.,Department of Medicine, Case Western Reserve University, Cleveland, United States.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, United States
| |
Collapse
|
18
|
Mazur-Bialy AI. Asprosin-A Fasting-Induced, Glucogenic, and Orexigenic Adipokine as a New Promising Player. Will It Be a New Factor in the Treatment of Obesity, Diabetes, or Infertility? A Review of the Literature. Nutrients 2021; 13:nu13020620. [PMID: 33673009 PMCID: PMC7918151 DOI: 10.3390/nu13020620] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/07/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Asprosin is a recently discovered protein released during fasting conditions mainly by adipocytes from white adipose tissue. As a glucogenic peptide, it stimulates the release of glucose from hepatic cells by binding to the OLFR734 receptor and leading to the activation of the G protein-cAMP-PKA pathway. As it crosses the blood–brain barrier, it also acts as an orexigenic peptide that stimulates food intake through activation of AgRP neurons in the hypothalamus; thus, asprosin participates in maintaining the body’s energy homeostasis. Moreover, studies have shown that asprosin levels are pathologically elevated in obesity and related diseases. However, the administration of anti-asprosin antibodies can both normalize its concentration and reduce food intake in obese mice, which makes it an interesting factor to combat obesity and related diseases. Current research also draws attention to the relationship between asprosin and fertility, especially in men. Asprosin improves age- and obesity-related decrease in fertility potential by improving sperm motility. It should also be mentioned that plasma asprosin levels can be differentially modulated by physical activity; intense anaerobic exercise increases asprosin level, while aerobic exercise decreases it. However, further research is necessary to confirm the exact mechanisms of asprosin activity and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Agnieszka Irena Mazur-Bialy
- Department of Biomechanics and Kinesiology, Faculty of Health Science, Institute of Physiotherapy, Jagiellonian University Medical College, Skawińska 8, 31-066 Krakow, Poland
| |
Collapse
|
19
|
Janoschek R, Hoffmann T, Morcos YAT, Sengle G, Dötsch J, Hucklenbruch-Rother E. Asprosin in pregnancy and childhood. Mol Cell Pediatr 2020; 7:18. [PMID: 33354737 PMCID: PMC7755955 DOI: 10.1186/s40348-020-00110-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/09/2020] [Indexed: 12/21/2022] Open
Affiliation(s)
- Ruth Janoschek
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany.
| | - Thorben Hoffmann
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany.
| | - Yousef Ashraf Tawfik Morcos
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany.,Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gerhard Sengle
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany.,Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Eva Hucklenbruch-Rother
- Department of Pediatrics, and Adolescent Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
20
|
A state of the art review on the novel mediator asprosin in the metabolic syndrome. Porto Biomed J 2020; 5:e108. [PMID: 33324783 PMCID: PMC7732265 DOI: 10.1097/j.pbj.0000000000000108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 10/08/2020] [Indexed: 01/06/2023] Open
Abstract
Metabolic syndrome is a complex and heterogeneous pathology characterized by a cluster of biochemical, clinical, and metabolic factors that came together in raising the risk of cardiovascular diseases, type 2 diabetes mellitus, and all-cause mortality. Some of these features are well defined in this syndrome like: obesity, inflammation, hypertension, insulin resistance, atherosclerotic dyslipidemias, endothelial dysfunction, and inflammation. This circuit is intermediated by a complex network of hormones, cytokines, transcription factors, and adipokines, among others. Some like leptin, adiponectin, Plasminogen activator inhibitor-1, interleukin-6, Tumor necrosis factor, and their influence on the metabolic syndrome are well described in the literature and new players are described continuously. One novel player was described in 2016 by Romere et al as a fasting-induced glycogenic protein hormone named asprosin. In order to perform a state-of-the-art, nonsystematic review of asprosin, a study of the available literature was carried out in the main database (Pubmed) and the results were studied and correlated to better understand the mechanism of action of this hormone. Asprosin is not only associated with the metabolic syndrome features like glucose and lipid metabolism, insulin resistance, obesity and inflammation but also in other pathologies metabolic syndrome related like diabetic retinopathy, polycystic ovary syndrome and anorexia nervosa. A limited number of pathways were already unveiled although much more research is needed to better understand the therapeutical potential of asprosin in the metabolic syndrome.
Collapse
|
21
|
Fibrillin-1 and fibrillin-1-derived asprosin in adipose tissue function and metabolic disorders. J Cell Commun Signal 2020; 14:159-173. [PMID: 32279186 DOI: 10.1007/s12079-020-00566-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/28/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
The extracellular matrix microenvironment of adipose tissue is of critical importance for the differentiation, remodeling and function of adipocytes. Fibrillin-1 is one of the main components of microfibrils and a key player in this process. Furin processing of profibrillin-1 results in mature fibrillin-1 and releases the C-terminal propeptide as a circulating hunger hormone, asprosin. Mutations in the fibrillin-1 gene lead to adipose tissue dysfunction and causes Marfan syndrome, marfanoid progeroid lipodystrophy syndrome, and neonatal progeroid syndrome. Increased TGF-β signaling, altered mechanical properties and impaired adipogenesis are potential causes of adipose tissue dysfunction, mediated through deficient microfibrils. Circulating asprosin on the other hand is secreted primarily by white adipose tissue under fasting conditions and in obesity. It increases hepatic glucose production and drives insulin secretion and appetite stimulation through inter-organ cross talk. This review discusses the metabolic consequences of fibrillin-1 and fibrillin-1-derived asprosin in pathological conditions. Understanding the dynamic role of fibrillin-1 in the adipose tissue milieu and of circulating asprosin in the body can provide novel mechanistic insights into how fibrillin-1 may contribute to metabolic syndrome. This could lead to new management regimens of patients with metabolic disease.
Collapse
|
22
|
Hoffmann JG, Xie W, Chopra AR. Energy Regulation Mechanism and Therapeutic Potential of Asprosin. Diabetes 2020; 69:559-566. [PMID: 32198197 PMCID: PMC7085243 DOI: 10.2337/dbi19-0009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 01/15/2020] [Indexed: 12/11/2022]
Abstract
Genetic studies of patients with neonatal progeroid syndrome led to the discovery of the novel fasting-induced, glucogenic, and orexigenic hormone named asprosin, the C-terminal cleavage product of profibrillin. Upon secretion, asprosin travels to the liver, where it exerts a glucogenic effect through OR4M1, an olfactory G-protein-coupled receptor. It also crosses the blood-brain barrier to stimulate appetite-modulating neurons in the arcuate nucleus of the hypothalamus, exerting an orexigenic effect via an as yet unidentified receptor. Specifically, it stimulates appetite by activating orexigenic AgRP neurons and inhibiting anorexigenic POMC neurons. Studies have also focused on the therapeutic potential of inhibiting asprosin for treatment of obesity and type 2 diabetes, both of which are characterized by high levels of circulating asprosin. It has been shown that anti-asprosin monoclonal antibodies reduce blood glucose, appetite, and body weight, validating asprosin as a therapeutic target. Current work aims to uncover key features of the asprosin biology such as the identification of its neuronal receptor, identification of the secretion mechanism from adipose tissue, and development of anti-asprosin monoclonal antibodies as diabetes and obesity therapies.
Collapse
Affiliation(s)
| | - Wei Xie
- Harrington Discovery Institute at University Hospitals, Cleveland, OH
| | - Atul R Chopra
- Harrington Discovery Institute at University Hospitals, Cleveland, OH
- Department of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
23
|
Yuan M, Li W, Zhu Y, Yu B, Wu J. Asprosin: A Novel Player in Metabolic Diseases. Front Endocrinol (Lausanne) 2020; 11:64. [PMID: 32153505 PMCID: PMC7045041 DOI: 10.3389/fendo.2020.00064] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 01/31/2020] [Indexed: 12/20/2022] Open
Abstract
Asprosin, a novel glucogenic adipokine, is encoded by two exons (exon 65 and exon 66) of the gene Fibrillin 1 (FBN1) and mainly synthesized and released by white adipose tissue during fasting. Asprosin plays a complex role in the central nervous system (CNS), peripheral tissues, and organs. It is involved in appetite, glucose metabolism, insulin resistance (IR), cell apoptosis, etc. In this review, we will summarize the newly discovered roles of asprosin in metabolic diseases including diabetes, obesity, polycystic ovarian syndrome (PCOS), and cardiovascular disease (CVD), which may contribute to future clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Mingyang Yuan
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Weidong Li
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Zhu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Boyao Yu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Wu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Research Center for Obesity and Its Metabolic Complications, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Jing Wu
| |
Collapse
|