1
|
Simonian TL, Meyer AS, Guo J, Sha J, Wohlschlegel JA, Droujinine IA, Perrimon N, McMahon AP. Sex and Depot Specific Adipocyte Proteome Profiling In Vivo via Intracellular Proximity Labeling. Compr Physiol 2025; 15:e70007. [PMID: 40181252 PMCID: PMC11969033 DOI: 10.1002/cph4.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025]
Abstract
Adipose tissue has varying distributions and metabolic properties between the sexes. Inherent sex-specific differences in adipocytes may heighten the risk of metabolic disease in males. Analysis of the adipocyte proteome can potentially provide important insight. To enable cell-type specific proteomic profiling in vivo, we genetically engineered a mouse line for cell-type specific production of a promiscuous biotin ligase (BirA*G3) facilitating the rapid isolation of biotinylated cell-type specific proteomes. Adipocyte-specific activation of cytoplasmic BirA*G3 led to robust biotinylation of adipocyte proteins across all major fat depots. Comparison of brown adipose tissue (BAT) and subcutaneous white adipose tissue (SAT) proteomes identified 229 brown adipose-enriched and 35 white adipose-enriched proteins. Regional comparison of white fat depots revealed additional differences across depots. Comparison of male and female depots identified sexually dimorphic adipose proteins: AHNAK predominating in the male and ACOT2 in the female. These findings validate the genetic model and highlight insights to be gained through targeted profiling of adipocytes. The genetic tool adds to existing approaches for in vivo proximity profiling of cell-type specific proteome programs.
Collapse
Affiliation(s)
- Taylor L. Simonian
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Amanda S. Meyer
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Jihui Sha
- Department of Biological Chemistry, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - James A. Wohlschlegel
- Department of Biological Chemistry, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Ilia A. Droujinine
- Department of Molecular MedicineScripps Research InstituteLa JollaCaliforniaUSA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik InstituteHarvard Medical SchoolCambridgeMassachusettsUSA
- Howard Hughes Medical InstituteChevy ChaseMarylandUSA
| | - Andrew P. McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
2
|
Setoyama D, Han D, Tian J, Lee H, Shin H, Nga H, Nguyen T, Moon J, Jang H, Kim E, Choe S, Ju S, Choi D, Kwon O, Yi H. Comparative Analysis of Primary Sarcopenia and End-Stage Renal Disease-Related Muscle Wasting Using Multi-Omics Approaches. J Cachexia Sarcopenia Muscle 2025; 16:e13749. [PMID: 40207397 PMCID: PMC11982700 DOI: 10.1002/jcsm.13749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/16/2024] [Accepted: 01/29/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Age-related primary sarcopenia and end-stage renal disease (ESRD)-related muscle wasting are discrete entities; however, both manifest as a decline in skeletal muscle mass and strength. The etiological pathways differ, with aging factors implicated in sarcopenia and a combination of uremic factors, including haemodialysis, contributing to ESRD-related muscle wasting. Understanding these molecular nuances is imperative for targeted interventions, and the integration of proteomic and metabolomic data elucidate these intricate processes. METHODS We generated detailed clinical data and multi-omics data (plasma proteomics and metabolomics) for 78 participants to characterise sarcopenia (n = 28; mean age, 72.6 ± 7.0 years) or ESRD (n = 22; 61.6 ± 5.5 years) compared with controls (n = 28; 69.3 ± 5.7 years). Muscle mass was measured using bioelectrical impedance analysis and handgrip strength. Five-times sit-to-stand test performance was measured for all participants. Sarcopenia was diagnosed in accordance with the 2019 Consensus Guidelines from the Asian Working Group for Sarcopenia. An abundance of 234 metabolites and 722 protein groups was quantified in all plasma samples using liquid chromatography with tandem mass spectrometry. RESULTS Muscle mass, handgrip strength and lower limb muscle function significantly lower in the sarcopenia group and the ESRD group compared with those in the control group. Metabolomics revealed altered metabolites, highlighting exclusive differences in ESRD-related muscle wasting. Metabolite set enrichment analysis revealed the involvement of numerous metabolic intermediates associated with urea cycle, amino acid metabolism and nucleic acid metabolism. Catecholamines, including epinephrine, dopamine and serotonin, are significantly elevated in the plasma of patients within the ESRD group. Proteomics data exhibited a clearer distinction among the three groups compared with the metabolomics data, particularly in distinguishing the control group from the sarcopenia group. The ciliary neurotrophic factor receptor was top-ranked in terms of the variable importance of projection scores. Plasma AHNAK protein levels was higher in the sarcopenia group but was lower in the ESRD group. Proteomic set enrichment analysis revealed enrichment of several pathways related to sarcopenia, such as hemopexin, defence response and cell differentiation, in sarcopenia group. Multi-omic integration analysis revealed associations between relevant metabolites, including catecholamines, and a group of annotated proteins in extracellular exosomes. CONCLUSIONS We identified distinct multi-omic signatures in individuals with ESRD or sarcopenia, providing new insights into the mechanisms underlying ESRD-related muscle wasting, which differ from primary sarcopenia. These findings may support interventions for context-dependent muscle loss and contribute to the development of targeted treatments and preventive strategies for muscle wasting.
Collapse
Affiliation(s)
- Daiki Setoyama
- Department of Clinical Chemistry and Laboratory MedicineKyushu University HospitalFukuokaJapan
| | - Dohyun Han
- Proteomics Core FacilitySeoul National University Hospital Biomedical Research InstituteSeoulSouth Korea
| | - Jingwen Tian
- Laboratory of Endocrinology and Immune SystemChungnam National University School of MedicineDaejeonSouth Korea
- Department of Medical ScienceChungnam National University School of MedicineDaejeonSouth Korea
| | - Ho Yeop Lee
- Laboratory of Endocrinology and Immune SystemChungnam National University School of MedicineDaejeonSouth Korea
- Department of Medical ScienceChungnam National University School of MedicineDaejeonSouth Korea
| | - Hyun Suk Shin
- Proteomics Core FacilitySeoul National University Hospital Biomedical Research InstituteSeoulSouth Korea
| | - Ha Thi Nga
- Laboratory of Endocrinology and Immune SystemChungnam National University School of MedicineDaejeonSouth Korea
- Department of Medical ScienceChungnam National University School of MedicineDaejeonSouth Korea
| | - Thi Linh Nguyen
- Laboratory of Endocrinology and Immune SystemChungnam National University School of MedicineDaejeonSouth Korea
- Department of Medical ScienceChungnam National University School of MedicineDaejeonSouth Korea
| | - Ji Sun Moon
- Laboratory of Endocrinology and Immune SystemChungnam National University School of MedicineDaejeonSouth Korea
| | - Hyo Ju Jang
- Laboratory of Endocrinology and Immune SystemChungnam National University School of MedicineDaejeonSouth Korea
- Department of Medical ScienceChungnam National University School of MedicineDaejeonSouth Korea
| | - Evonne Kim
- Department of Biomedical Sciences, BK21 FOUR Biomedical Science ProgramSeoul National University College of MedicineSeoulSouth Korea
| | - Seong‐Kyu Choe
- Department of Medicine, Graduate SchoolWonkwang UniversityIksanSouth Korea
- Sarcopenia Total Solution CenterWonkwang UniversityIksanSouth Korea
| | - Sang Hyeon Ju
- Department of Internal MedicineChungnam National University School of MedicineDaejeonSouth Korea
| | - Dae Eun Choi
- Department of Internal MedicineChungnam National University School of MedicineDaejeonSouth Korea
| | - Obin Kwon
- Department of Biomedical Sciences, BK21 FOUR Biomedical Science ProgramSeoul National University College of MedicineSeoulSouth Korea
- Department of Biochemistry and Molecular BiologySeoul National University College of MedicineSeoulSouth Korea
- Genomic Medicine Institute, Medical Research CenterSeoul National UniversitySeoulSouth Korea
| | - Hyon‐Seung Yi
- Laboratory of Endocrinology and Immune SystemChungnam National University School of MedicineDaejeonSouth Korea
- Department of Medical ScienceChungnam National University School of MedicineDaejeonSouth Korea
- Sarcopenia Total Solution CenterWonkwang UniversityIksanSouth Korea
- Department of Internal MedicineChungnam National University School of MedicineDaejeonSouth Korea
| |
Collapse
|
3
|
Xu Y, Huang L, Zhuang Y, Huang H. Modulation of adipose tissue metabolism by exosomes in obesity. Am J Physiol Endocrinol Metab 2024; 326:E709-E722. [PMID: 38416071 DOI: 10.1152/ajpendo.00155.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 02/14/2024] [Accepted: 02/17/2024] [Indexed: 02/29/2024]
Abstract
Obesity and its related metabolic complications represent a significant global health challenge. Central to this is the dysregulation of glucolipid metabolism, with a predominant focus on glucose metabolic dysfunction in the current research, whereas adipose metabolism impairment garners less attention. Exosomes (EXs), small extracellular vesicles (EVs) secreted by various cells, have emerged as important mediators of intercellular communication and have the potential to be biomarkers, targets, and therapeutic tools for diverse diseases. In particular, EXs have been found to play a role in adipose metabolism by transporting cargoes such as noncoding RNAs (ncRNA), proteins, and other factors. This review article summarizes the current understanding of the role of EXs in mediating adipose metabolism disorders in obesity. It highlights their roles in adipogenesis (encompassing adipogenic differentiation and lipid synthesis), lipid catabolism, lipid transport, and white adipose browning. The insights provided by this review offer new avenues for developing exosome-based therapies to treat obesity and its associated comorbidities.
Collapse
Affiliation(s)
- Yajing Xu
- Department of Endocrinology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, People's Republic of China
| | - Linghong Huang
- Department of Endocrinology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, People's Republic of China
| | - Yong Zhuang
- Department of Endocrinology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, People's Republic of China
| | - Huibin Huang
- Department of Endocrinology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, People's Republic of China
| |
Collapse
|
4
|
Zhang S, Cai Z, Li H. AHNAKs roles in physiology and malignant tumors. Front Oncol 2023; 13:1258951. [PMID: 38033502 PMCID: PMC10682155 DOI: 10.3389/fonc.2023.1258951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
The AHNAK family currently consists of two members, namely AHNAK and AHNAK2, both of which have a molecular weight exceeding 600 kDa. Homologous sequences account for approximately 90% of their composition, indicating a certain degree of similarity in terms of molecular structure and biological functions. AHNAK family members are involved in the regulation of various biological functions, such as calcium channel modulation and membrane repair. Furthermore, with advancements in biological and bioinformatics technologies, research on the relationship between the AHNAK family and tumors has rapidly increased in recent years, and its regulatory role in tumor progression has gradually been discovered. This article briefly describes the physiological functions of the AHNAK family, and reviews and analyzes the expression and molecular regulatory mechanisms of the AHNAK family in malignant tumors using Pubmed and TCGA databases. In summary, AHNAK participates in various physiological and pathological processes in the human body. In multiple types of cancers, abnormal expression of AHNAK and AHNAK2 is associated with prognosis, and they play a key regulatory role in tumor progression by activating signaling pathways such as ERK, MAPK, Wnt, and MEK, as well as promoting epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Shusen Zhang
- Hebei Province Xingtai People’s Hospital Postdoctoral Workstation, Xingtai, China
- Postdoctoral Mobile Station, Hebei Medical University, Shijiazhuang, China
- Department of Pulmonary and Critical Care Medicine, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, China
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhigang Cai
- Postdoctoral Mobile Station, Hebei Medical University, Shijiazhuang, China
- The First Department of Pulmonary and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hui Li
- Department of surgery, Affiliated Xing Tai People Hospital of Hebei Medical University, Xingtai, China
| |
Collapse
|
5
|
Zhou H, Chen C, Hu H, Jiang B, Yin Y, Zhang K, Shen M, Wu S, Wang Z. High-intensity interval training improves fatty infiltration in the rotator cuff through the β3 adrenergic receptor in mice. Bone Joint Res 2023; 12:455-466. [PMID: 37524338 PMCID: PMC10390263 DOI: 10.1302/2046-3758.128.bjr-2022-0309.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/02/2023] Open
Abstract
Aims Rotator cuff muscle atrophy and fatty infiltration affect the clinical outcomes of rotator cuff tear patients. However, there is no effective treatment for fatty infiltration at this time. High-intensity interval training (HIIT) helps to activate beige adipose tissue. The goal of this study was to test the role of HIIT in improving muscle quality in a rotator cuff tear model via the β3 adrenergic receptor (β3AR). Methods Three-month-old C57BL/6 J mice underwent a unilateral rotator cuff injury procedure. Mice were forced to run on a treadmill with the HIIT programme during the first to sixth weeks or seventh to 12th weeks after tendon tear surgery. To study the role of β3AR, SR59230A, a selective β3AR antagonist, was administered to mice ten minutes before each exercise through intraperitoneal injection. Supraspinatus muscle, interscapular brown fat, and inguinal subcutaneous white fat were harvested at the end of the 12th week after tendon tear and analyzed biomechanically, histologically, and biochemically. Results Histological analysis of supraspinatus muscle showed that HIIT improved muscle atrophy, fatty infiltration, and contractile force compared to the no exercise group. In the HIIT groups, supraspinatus muscle, interscapular brown fat, and inguinal subcutaneous white fat showed increased expression of tyrosine hydroxylase and uncoupling protein 1, and upregulated the β3AR thermogenesis pathway. However, the effect of HIIT was not present in mice injected with SR59230A, suggesting that HIIT affected muscles via β3AR. Conclusion HIIT improved supraspinatus muscle quality and function after rotator cuff tears by activating systemic sympathetic nerve fibre near adipocytes and β3AR.
Collapse
Affiliation(s)
- Hecheng Zhou
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
- Xiangya Medical School of Central South University, Changsha, China
| | - Chuanshun Chen
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
- Xiangya Medical School of Central South University, Changsha, China
| | - Hai Hu
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Binbin Jiang
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yuesong Yin
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
- Xiangya Medical School of Central South University, Changsha, China
| | - Kexiang Zhang
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Minren Shen
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Song Wu
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zili Wang
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
6
|
Zhou H, Wang Z, Chen C, Hu H, Jiang B, Yin Y, Zhang K, Shen M, Wu S. Effect of High-Intensity Interval Training on Fatty Infiltration After Delayed Rotator Cuff Repair in a Mouse Model. Orthop J Sports Med 2023; 11:23259671231170192. [PMID: 37223073 PMCID: PMC10201644 DOI: 10.1177/23259671231170192] [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: 01/26/2023] [Accepted: 02/15/2023] [Indexed: 05/25/2023] Open
Abstract
Background Fatty infiltration (FI) of the rotator cuff muscles is correlated with shoulder function and retear rates after rotator cuff repair. High-intensity interval training (HIIT) induces beige adipose tissue to express more uncoupling protein 1 (UCP1) to consume lipids. The beta-3 adrenergic receptor (β3AR) is located on adipocyte membrane and induces thermogenesis. Purpose To test the role of HIIT in improving muscle quality and contractility in a delayed rotator cuff repair mouse model via β3AR. Study Design Controlled laboratory study. Methods Three-month-old C57BL/6J mice underwent a unilateral supraspinatus (SS) tendon transection with a 6-week delayed tendon repair. Mice ran on a treadmill with the HIIT program for 6 weeks after tendon transection or after delayed repair. To study the role of β3AR, SR59230A, a selective β3AR antagonist, was administered to mice 10 minutes before each exercise through intraperitoneal injection. The SS, interscapular brown adipose tissue (iBAT), and subcutaneous inguinal white adipose tissue (ingWAT) were harvested at the end of the 12th week after tendon transection and were analyzed by histology and Western blotting. Tests were performed to assess muscle contractility of the SS. Results Histologic analysis of SS showed that HIIT prevented and reversed muscle atrophy and FI. The contractile tests showed higher contractility of the SS in the HIIT groups than in the no-exercise group. In the HIIT groups, SS, iBAT, and ingWAT all showed increased expression of tyrosine hydroxylase, UCP1, and upregulated β3AR thermogenesis pathway. However, SR59230A inhibited HIIT, suggesting that the effect of HIIT depends on β3AR. Conclusion HIIT improved SS quality and function after delayed rotator cuff repair through a β3AR-dependent mechanism. Clinical Relevance HIIT may serve as a new rehabilitation method for patients with rotator cuff muscle atrophy and FI after rotator cuff repair to improve postoperative clinical outcomes.
Collapse
Affiliation(s)
- Hecheng Zhou
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
- Clinical Medicine Eight-Year Program, Xiangya Medical School of Central South University, Changsha, Hunan, China
| | - Zili Wang
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Chuanshun Chen
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
- Clinical Medicine Eight-Year Program, Xiangya Medical School of Central South University, Changsha, Hunan, China
| | - Hai Hu
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Binbin Jiang
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Yuesong Yin
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
- Clinical Medicine Eight-Year Program, Xiangya Medical School of Central South University, Changsha, Hunan, China
| | - Kexiang Zhang
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Minren Shen
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Song Wu
- Department of Orthopaedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| |
Collapse
|
7
|
Osuna-Prieto FJ, Martinez-Tellez B, Segura-Carretero A, Ruiz JR. Activation of Brown Adipose Tissue and Promotion of White Adipose Tissue Browning by Plant-based Dietary Components in Rodents: A Systematic Review. Adv Nutr 2021; 12:2147-2156. [PMID: 34265040 PMCID: PMC8634450 DOI: 10.1093/advances/nmab084] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/30/2021] [Accepted: 06/15/2021] [Indexed: 12/21/2022] Open
Abstract
Activation of brown adipose tissue (BAT) and promotion of white adipose tissue (WAT) browning is considered a potential tool to combat obesity and cardiometabolic disorders. The use of plant-based dietary components has become one of the most used strategies for activating BAT and promoting WAT browning in rodents. The main reason is because plant-based dietary components are usually recognized as safe when the dose is properly adjusted, and they can easily be administrated by being added to the diet or dissolved in water. The present systematic review aimed to study the effects of plant-based dietary components on activation of BAT and promotion of WAT browning in rodents. A systematic search of PubMed and Scopus (from 1978 to 2019) identified eligible studies. Studies assessing the effects of plant-based dietary components added to diet and/or water on uncoupling protein 1 (UCP1) expression in BAT and/or WAT were included. Studies that used dietary components of animal origin, did not specify the effects on UCP1, or were conducted in other species different from mice or rats were excluded. Of 3919 studies identified in the initial screening, 146 studies were finally included in the review. We found that tea extract catechins, resveratrol, capsaicin and capsinoids, cacao extract flavanols, and quercetin were the most studied components. Scientific evidence suggests that some of these dietary components activate BAT and promote WAT browning via activation of the AMP-activated protein kinase (AMPK) and sirtuin 1 (SIRT1) pathways. These findings reveal that there is strong scientific evidence supporting the use of plant-based dietary components to activate BAT and promote WAT browning in rodents and thus to potentially combat obesity and cardiometabolic disorders.
Collapse
Affiliation(s)
| | - Borja Martinez-Tellez
- PROFITH (PROmoting FITness and Health through Physical Activity) Research Group, Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain,Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Antonio Segura-Carretero
- Department of Analytical Chemistry, University of Granada, Granada, Spain,Research and Development of Functional Food Centre (CIDAF), Health Science Technological Park Avda. Del Conocimiento, Granada, Spain
| | - Jonatan R Ruiz
- PROFITH (PROmoting FITness and Health through Physical Activity) Research Group, Department of Physical Education and Sport, Faculty of Sport Sciences, University of Granada, Granada, Spain
| |
Collapse
|
8
|
Ahnak deficiency attenuates high-fat diet-induced fatty liver in mice through FGF21 induction. Exp Mol Med 2021; 53:468-482. [PMID: 33785868 PMCID: PMC8080712 DOI: 10.1038/s12276-021-00573-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 02/01/2023] Open
Abstract
The AHNAK nucleoprotein has been determined to exert an anti-obesity effect in adipose tissue and further inhibit adipogenic differentiation. In this study, we examined the role of AHNAK in regulating hepatic lipid metabolism to prevent diet-induced fatty liver. Ahnak KO mice have reportedly exhibited reduced fat accumulation in the liver and decreased serum triglyceride (TG) levels when provided with either a normal chow diet or a high-fat diet (HFD). Gene expression profiling was used to identify novel factors that could be modulated by genetic manipulation of the Ahnak gene. The results revealed that fibroblast growth factor 21 (FGF21) was markedly increased in the livers of Ahnak KO mice compared with WT mice fed a HFD. Ahnak knockdown in hepatocytes reportedly prevented excessive lipid accumulation induced by palmitate treatment and was associated with increased secretion of FGF21 and the expression of genes involved in fatty acid oxidation, which are primarily downstream of PPARα. These results indicate that pronounced obesity and hepatic steatosis are attenuated in HFD-fed Ahnak KO mice. This may be attributed, in part, to the induction of FGF21 and regulation of lipid metabolism, which are considered to be involved in increased fatty acid oxidation and reduced lipogenesis in the liver. These findings suggest that targeting AHNAK may have beneficial implications in preventing or treating hepatic steatosis.
Collapse
|
9
|
Im H, Park JH, Im S, Han J, Kim K, Lee YH. Regulatory roles of G-protein coupled receptors in adipose tissue metabolism and their therapeutic potential. Arch Pharm Res 2021; 44:133-145. [PMID: 33550564 PMCID: PMC7907040 DOI: 10.1007/s12272-021-01314-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022]
Abstract
The high incidence of obesity has increased the need to discover new therapeutic targets to combat obesity and obesity-related metabolic diseases. Obesity is defined as an abnormal accumulation of adipose tissue, which is one of the major metabolic organs that regulate energy homeostasis. However, there are currently no approved anti-obesity therapeutics that directly target adipose tissue metabolism. With recent advances in the understanding of adipose tissue biology, molecular mechanisms involved in brown adipose tissue expansion and metabolic activation have been investigated as potential therapeutic targets to increase energy expenditure. This review focuses on G-protein coupled receptors (GPCRs) as they are the most successful class of druggable targets in human diseases and have an important role in regulating adipose tissue metabolism. We summarize recent findings on the major GPCR classes that regulate thermogenesis and mitochondrial metabolism in adipose tissue. Improved understanding of GPCR signaling pathways that regulate these processes could facilitate the development of novel pharmacological approaches to treat obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Hyeonyeong Im
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University; Bio-MAX Institute, Seoul National University, 29-Room # 311, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Ji-Hyun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University; Bio-MAX Institute, Seoul National University, 29-Room # 311, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Seowoo Im
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University; Bio-MAX Institute, Seoul National University, 29-Room # 311, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Juhyeong Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University; Bio-MAX Institute, Seoul National University, 29-Room # 311, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Kyungmin Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University; Bio-MAX Institute, Seoul National University, 29-Room # 311, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Yun-Hee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University; Bio-MAX Institute, Seoul National University, 29-Room # 311, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
10
|
The S100B Protein and Partners in Adipocyte Response to Cold Stress and Adaptive Thermogenesis: Facts, Hypotheses, and Perspectives. Biomolecules 2020; 10:biom10060843. [PMID: 32486507 PMCID: PMC7356379 DOI: 10.3390/biom10060843] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/20/2020] [Accepted: 05/29/2020] [Indexed: 12/17/2022] Open
Abstract
In mammals, adipose tissue is an active secretory tissue that responds to mild hypothermia and as such is a genuine model to study molecular and cellular adaptive responses to cold-stress. A recent study identified a mammal-specific protein of the endoplasmic reticulum that is strongly induced in the inguinal subcutaneous white adipocyte upon exposure to cold, calsyntenin 3β (CLSTN3β). CLSTN3β regulates sympathetic innervation of thermogenic adipocytes and contributes to adaptive non-shivering thermogenesis. The calcium- and zinc-binding S100B is a downstream effector in the CLSTN3β pathways. We review, here, the literature on the transcriptional regulation of the S100b gene in adipocyte cells. We also rationalize the interactions of the S100B protein with its recognized or hypothesized intracellular (p53, ATAD3A, CYP2E1, AHNAK) and extracellular (Receptor for Advanced Glycation End products (RAGE), RPTPσ) target proteins in the context of adipocyte differentiation and adaptive thermogenesis. We highlight a chaperon-associated function for the intracellular S100B and point to functional synergies between the different intracellular S100B target proteins. A model of non-classical S100B secretion involving AHNAK/S100A10/annexin2-dependent exocytosis by the mean of exosomes is also proposed. Implications for related areas of research are noted and suggestions for future research are offered.
Collapse
|
11
|
Luo Z, Zhang T, Wang S, He Y, Ye Q, Cao W. The Trp64Arg polymorphism in β3 adrenergic receptor (ADRB3) gene is associated with adipokines and plasma lipids: a systematic review, meta-analysis, and meta-regression. Lipids Health Dis 2020; 19:99. [PMID: 32430022 PMCID: PMC7236936 DOI: 10.1186/s12944-020-01290-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/15/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Recently, some studies claim that adipokines may modulate plasma lipids. More interestingly, the ADRB3 Trp64Arg polymorphism may regulate adipokines and play an essential role in lipids metabolism. This study aims to clarify the associations of ADRB3 Trp64Arg polymorphism with plasma adipokines and lipid levels. METHODS Twenty-two studies (5527 subjects) and 121 studies (54,059 subjects) were respectively identified for the association analyses of adipokines and lipids. Standardized mean difference (SMD) and 95% confidence interval (CI) were used to estimate the strength of the Trp64Arg variant in adipokines and plasma lipids. All results were recalculated after eliminating the studies with heterogeneity. RESULTS The carriers of the C allele (Arg at 64th position was encoded by the C allele) had higher levels of leptin and lower levels of adiponectin than the non-carriers. The carriers of the C allele had higher levels of triglycerides (TG), total cholesterol (TC), and lower levels of high-density lipoprotein cholesterol (HDL-C) than the non-carriers. Subgroup analysis certified an ethnicity (Asians), disease status (obesity), and gender (females) specific association. Sensitivity analysis indicated that the analysis results were robust and stable. Meta-regression indicated that obesity was related to adiponectin. CONCLUSIONS The C allele carriers of Trp64Arg polymorphism had a slight but significant influence on lipid levels, and the remarkable effects specific existed in obese Asian women. The associations of Trp64Arg polymorphism with dyslipidemia may partly be mediated by the effect of this polymorphism on adipokines. The association of Trp64Arg polymorphism with obesity may partly be mediated by the effect of this polymorphism on adipokines. The C allele carriers had abnormal levels of adipokines and lipids, and it indicated that the Trp64Arg polymorphism might represent a genetic risk factor for coronary artery disease (CAD).
Collapse
Affiliation(s)
- Zhi Luo
- Department of Cardiology, The First People's Hospital of Zigong, Zigong, 643000, People's Republic of China.
| | - Ting Zhang
- Department of Nursing, Sichuan Vocational College of Health and Rehabilitation, Zigong, 643000, People's Republic of China
| | - Shengping Wang
- North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Yuxian He
- North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Qiutang Ye
- Office of Research Affairs, The First People's Hospital of Zigong, Zigong, 643000, People's Republic of China
| | - Wenzhai Cao
- Department of Cardiology, The First People's Hospital of Zigong, Zigong, 643000, People's Republic of China.
| |
Collapse
|
12
|
Genes dysregulated in the blood of people with Williams syndrome are enriched in protein-coding genes positively selected in humans. Eur J Med Genet 2020; 63:103828. [DOI: 10.1016/j.ejmg.2019.103828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/09/2019] [Accepted: 12/21/2019] [Indexed: 12/29/2022]
|
13
|
Intensive morphometric analysis of enormous alterations in skeletal bone system with micro-CT for AHNAK -/- mice. Anat Sci Int 2020; 95:323-333. [PMID: 32067190 DOI: 10.1007/s12565-020-00525-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 01/14/2020] [Indexed: 10/25/2022]
Abstract
AHNAK has been reported to be involved in actin cytoskeleton rearrangement of some cell types, calcium homeostasis, and activation of T cells. Although the functional role of AHNAK in muscle cells, epidermis, and brain has been determined, its association with apparent clinical impairment has not been found yet. During phenotypic analysis of AHNAK knock out (KO) mice for many years, we observed that AHNAK KO mice showed very slow growth. Snouts of these animals were very short, and their bones were easily broken compared to normal mice. It is known that AHNAK is closely related to calcium. However, intensive morphological studies on phenotypes of bone have yet been reported for AHNAK. Thus, the objective of the present study was to analyze the morphology of skull, mandibular, limbs, and caudal bones of AHNAK KO mice intensively using micro-CT with many factors for various ages of these mice (6 weeks, 18 weeks, and 40 weeks). As a result, it was found that the facial region of AHNAK KO mouse showed a large difference in mandible than skull. Their both femur and tibia were shortened, and bone strength was also significantly decreased compared to normal mice. Particularly, the tail bone of AHNAK KO mice exhibited morphological abnormality by age. Taken together, these results suggest that AHNAK plays an important role in bone shape, development, and metabolism. Although our results demonstrated that AHNAK has a distinct role in bone, further investigations are needed to determine various features of bone metabolism related to AHNAK in the future.
Collapse
|
14
|
|
15
|
Choi EW, Lee HW, Lee JS, Kim IY, Shin JH, Seong JK. Ahnak-knockout mice show susceptibility to Bartonella henselae infection because of CD4+ T cell inactivation and decreased cytokine secretion. BMB Rep 2019. [PMID: 30940323 PMCID: PMC6507843 DOI: 10.5483/bmbrep.2019.52.4.310] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The present study evaluated the role of AHNAK in Bartonella henselae infection. Mice were intraperitoneally inoculated with 2 × 108 colony-forming units of B. henselae Houston-1 on day 0 and subsequently on day 10. Blood and tissue samples of the mice were collected 8 days after the final B. henselae injection. B. henselae infection in the liver of Ahnak-knockout and wild-type mice was confirmed by performing polymerase chain reaction, with Bartonella adhesion A as a marker. The proportion of B. henselae-infected cells increased in the liver of the Ahnak-knockout mice. Granulomatous lesions, inflammatory cytokine levels, and liver enzyme levels were also higher in the liver of the Ahnak-knockout mice than in the liver of the wild-type mice, indicating that Ahnak deletion accelerated B. henselae infection. The proportion of CD4+interferon-γ (IFN-γ)+ and CD4+interleukin (IL)-4+ cells was significantly lower in the B. henselae-infected Ahnak-knockout mice than in the B. henselae-infected wild-type mice. In vitro stimulation with B. henselae significantly increased IFN-γ and IL-4 secretion in the splenocytes obtained from the B. henselae-infected wild-type mice, but did not increase IFN-γ and IL-4 secretion in the splenocytes obtained from the B. henselae-infected Ahnak-KO mice. In contrast, IL-1α, IL-1β, IL-6, IL-10, RANTES, and tumor necrosis factor-α secretion was significantly elevated in the splenocytes obtained from both B. henselae-infected wild-type and Ahnak-knockout mice. These results indicate that Ahnak deletion promotes B. henselae infection. Impaired IFN-γ and IL-4 secretion in the Ahnak-knockout mice suggests the impairment of Th1 and Th2 immunity in these mice.
Collapse
Affiliation(s)
- Eun Wha Choi
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
- Laboratory Animal Research Center, Samsung Biomedical Research Institute, Samsung Medical Center, Seoul 06351, Korea
| | - Hee Woo Lee
- Institute of Research and Development, Chaon Corp., Seongnam 13493, Korea
| | - Jun Sik Lee
- Department of Biology, Immunology Research Lab., College of Natural Sciences, Chosun University, Gwangju 61452, Korea
| | - Il Yong Kim
- Laboratory of Developmental Biology and Genomics, BK21 Plus Program for Advanced Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, and Korea Mouse Phenotyp
| | - Jae Hoon Shin
- Laboratory of Developmental Biology and Genomics, BK21 Plus Program for Advanced Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, and Korea Mouse Phenotyp
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, BK21 Plus Program for Advanced Veterinary Science, Research Institute for Veterinary Science, College of Veterinary Medicine, and Korea Mouse Phenotyp
- Interdiscplinary Program for Bioinformatics, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
16
|
Chen L, Shi G, Chen G, Li J, Li M, Zou C, Fang C, Li C. Transcriptome Analysis Suggests the Roles of Long Intergenic Non-coding RNAs in the Growth Performance of Weaned Piglets. Front Genet 2019; 10:196. [PMID: 30936891 PMCID: PMC6431659 DOI: 10.3389/fgene.2019.00196] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/25/2019] [Indexed: 11/19/2022] Open
Abstract
Long intergenic non-coding RNAs (lincRNAs) have been considered to play a key regulatory role in various biological processes. An increasing number of studies have utilized transcriptome analysis to obtain lincRNAs with functions related to cancer, but lincRNAs affecting growth rates in weaned piglets are rarely described. Although lincRNAs have been systematically identified in various mouse tissues and cell lines, studies of lincRNA in pigs remain rare. Therefore, identifying and characterizing novel lincRNAs affecting the growth performance of weaned piglets is of great importance. Here, we reconstructed 101,988 lincRNA transcripts and identified 1,078 lincRNAs in two groups of longissimus dorsi muscle (LDM) and subcutaneous fat (SF) based on published RNA-seq datasets. These lincRNAs exhibit typical characteristics, such as shorter lengths and lower expression relative to protein-encoding genes. Gene ontology analysis revealed that some lincRNAs could be involved in weaned piglet related processes, such as insulin resistance and the AMPK signaling pathway. We also compared the positional relationship between differentially expressed lincRNAs (DELs) and quantitative trait loci (QTL) and found that some of DELs may play an important role in piglet growth and development. Our work details part of the lincRNAs that may affect the growth performance of weaned piglets and promotes future studies of lincRNAs for molecular-assisted development in weaned piglets.
Collapse
Affiliation(s)
- Lin Chen
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Gaoli Shi
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Guoting Chen
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jingxuan Li
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Mengxun Li
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Cheng Zou
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Chengchi Fang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Changchun Li
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
17
|
Essential role of Ahnak in adipocyte differentiation leading to the transcriptional regulation of Bmpr1α expression. Cell Death Dis 2018; 9:864. [PMID: 30154465 PMCID: PMC6113281 DOI: 10.1038/s41419-018-0873-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 06/08/2018] [Accepted: 07/09/2018] [Indexed: 01/05/2023]
Abstract
The role of Ahnak in obesity has been reported previously. Loss of Ahnak leads to decreased Bmp4/Smad1 signaling, resulting in the downregulation of adipocyte differentiation. However, the biological significance of Ahnak remains largely unknown. In this study, we demonstrate that Ahnak-mediated impaired adipogenesis results in decreased Bmpr1α transcriptional expression. To confirm this, Ahnak siRNA was used to knock-down Ahnak in C3H10T1/2 and primary stromal vascular fraction cells. Ahnak siRNA transfected cells showed suppression of Bmpr1α expression and decreased BMP4/ Bmpr1α signaling. The differential adipogenesis was further confirmed by knock-down of Bmpr1α in C3H10T1/2 cells, which resulted in reduced adipogenesis. Moreover, stable Ahnak knock-out C3H10T1/2 cells stably transfected with Ahnak CRISPR/Cas9 plasmid suppressed expression of Bmpr1α and prevented differentiation into adipocytes. Furthermore, we developed immortalized pre-adipocytes from wild-type or Ahnak Knock-out mice's stromal vascular fraction (SVF) to confirm the function of Ahnak in pre-adipocyte transition. Immortalized Ahnak knock-out SVF cells showed lower level of Bmpr1α expression, evidence by their impaired BMP4/Bmpr1α signaling. Upon adipogenic induction, immortalized Ahnak knock-out SVF cells exhibited a marked decrease in adipocyte differentiation compared with immortalized wild-type pre-adipocytes. Furthermore, over-expression of Bmpr1α restored the adipogenic activity of Ahnak knock-out C3H10T1/2 cells and immortalized Ahnak knock-out SVF cells. Our data reveal the missing link in Ahnak-mediated adipose tissue remodeling and suggest that precise regulation of Ahnak in adipose tissue might have a therapeutic advantage for metabolic disease treatment.
Collapse
|
18
|
Park JW, Kim IY, Choi JW, Lim HJ, Shin JH, Kim YN, Lee SH, Son Y, Sohn M, Woo JK, Jeong JH, Lee C, Bae YS, Seong JK. AHNAK Loss in Mice Promotes Type II Pneumocyte Hyperplasia and Lung Tumor Development. Mol Cancer Res 2018; 16:1287-1298. [PMID: 29724814 DOI: 10.1158/1541-7786.mcr-17-0726] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/24/2018] [Accepted: 04/19/2018] [Indexed: 11/16/2022]
Abstract
AHNAK is known to be a tumor suppressor in breast cancer due to its ability to activate the TGFβ signaling pathway. However, the role of AHNAK in lung tumor development and progression remains unknown. Here, the Ahnak gene was disrupted to determine its effect on lung tumorigenesis and the mechanism by which it triggers lung tumor development was investigated. First, AHNAK protein expression was determined to be decreased in human lung adenocarcinomas compared with matched nonneoplastic lung tissues. Then, Ahnak -/- mice were used to investigate the role of AHNAK in pulmonary tumorigenesis. Ahnak -/- mice showed increased lung volume and thicker alveolar walls with type II pneumocyte hyperplasia. Most importantly, approximately 20% of aged Ahnak -/- mice developed lung tumors, and Ahnak -/- mice were more susceptible to urethane-induced pulmonary carcinogenesis than wild-type mice. Mechanistically, Ahnak deficiency promotes the cell growth of lung epithelial cells by suppressing the TGFβ signaling pathway. In addition, increased numbers of M2-like alveolar macrophages (AM) were observed in Ahnak -/- lungs, and the depletion of AMs in Ahnak -/- lungs alleviated lung hyperplastic lesions, suggesting that M2-like AMs promoted the progression of lung hyperplastic lesions in Ahnak-null mice. Collectively, AHNAK suppresses type II pneumocyte proliferation and inhibits tumor-promoting M2 alternative activation of macrophages in mouse lung tissue. These results suggest that AHNAK functions as a novel tumor suppressor in lung cancer.Implications: The tumor suppressor function of AHNAK, in murine lungs, occurs by suppressing alveolar epithelial cell proliferation and modulating lung microenvironment. Mol Cancer Res; 16(8); 1287-98. ©2018 AACR.
Collapse
Affiliation(s)
- Jun Won Park
- Laboratory of Developmental Biology and Genomics, BK21 Program Plus for Advanced Veterinary Science, and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Korea.,Korea Mouse Phenotyping Center (KMPC), Seoul, Korea
| | - Il Yong Kim
- Laboratory of Developmental Biology and Genomics, BK21 Program Plus for Advanced Veterinary Science, and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Korea.,Korea Mouse Phenotyping Center (KMPC), Seoul, Korea
| | - Ji Won Choi
- Laboratory of Developmental Biology and Genomics, BK21 Program Plus for Advanced Veterinary Science, and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Hee Jung Lim
- Korea Mouse Phenotyping Center (KMPC), Seoul, Korea
| | - Jae Hoon Shin
- Laboratory of Developmental Biology and Genomics, BK21 Program Plus for Advanced Veterinary Science, and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Yo Na Kim
- Laboratory of Developmental Biology and Genomics, BK21 Program Plus for Advanced Veterinary Science, and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Seo Hyun Lee
- Laboratory of Developmental Biology and Genomics, BK21 Program Plus for Advanced Veterinary Science, and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Yeri Son
- Laboratory of Developmental Biology and Genomics, BK21 Program Plus for Advanced Veterinary Science, and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Mira Sohn
- Department of Life Sciences, Ewha Womans University, Seoul, Korea
| | - Jong Kyu Woo
- Laboratory of Developmental Biology and Genomics, BK21 Program Plus for Advanced Veterinary Science, and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Korea.,Korea Mouse Phenotyping Center (KMPC), Seoul, Korea
| | | | - Cheolju Lee
- Center for Theragnosis, Korea Institute of Science and Technology, Seoul, Korea
| | - Yun Soo Bae
- Department of Life Sciences, Ewha Womans University, Seoul, Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, BK21 Program Plus for Advanced Veterinary Science, and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Korea. .,Korea Mouse Phenotyping Center (KMPC), Seoul, Korea.,Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX/N-Bio Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
19
|
Yang J, Hagen J, Guntur KV, Allette K, Schuyler S, Ranjan J, Petralia F, Gesta S, Sebra R, Mahajan M, Zhang B, Zhu J, Houten S, Kasarskis A, Vishnudas VK, Akmaev VR, Sarangarajan R, Narain NR, Schadt EE, Argmann CA, Tu Z. A next generation sequencing based approach to identify extracellular vesicle mediated mRNA transfers between cells. BMC Genomics 2017; 18:987. [PMID: 29273013 PMCID: PMC5741891 DOI: 10.1186/s12864-017-4359-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 11/29/2017] [Indexed: 02/08/2023] Open
Abstract
Background Exosomes and other extracellular vesicles (EVs) have emerged as an important mechanism of cell-to-cell communication. However, previous studies either did not fully resolve what genetic materials were shuttled by exosomes or only focused on a specific set of miRNAs and mRNAs. A more systematic method is required to identify the genetic materials that are potentially transferred during cell-to-cell communication through EVs in an unbiased manner. Results In this work, we present a novel next generation of sequencing (NGS) based approach to identify EV mediated mRNA exchanges between co-cultured adipocyte and macrophage cells. We performed molecular and genomic profiling and jointly considered data from RNA sequencing (RNA-seq) and genotyping to track the “sequence varying mRNAs” transferred between cells. We identified 8 mRNAs being transferred from macrophages to adipocytes and 21 mRNAs being transferred in the opposite direction. These mRNAs represented biological functions including extracellular matrix, cell adhesion, glycoprotein, and signal peptides. Conclusions Our study sheds new light on EV mediated RNA communications between adipocyte and macrophage cells, which may play a significant role in developing insulin resistance in diabetic patients. This work establishes a new method that is applicable to examining genetic material exchanges in many cellular systems and has the potential to be extended to in vivo studies as well. Electronic supplementary material The online version of this article (10.1186/s12864-017-4359-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jialiang Yang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jacob Hagen
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Kimaada Allette
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sarah Schuyler
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Francesca Petralia
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Robert Sebra
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Milind Mahajan
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bin Zhang
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jun Zhu
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sander Houten
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Andrew Kasarskis
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | | | | | | | - Eric E Schadt
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Carmen A Argmann
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Zhidong Tu
- Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
20
|
Menezes TP, Hill E, de Alencar Moura A, Lobo MDP, Monteiro-Moreira ACO, Breton S, Machado-Neves M. Pattern of protein expression in the epididymis of Oligoryzomys nigripes (Cricetidae, Sigmodontinae). Cell Tissue Res 2017; 372:135-147. [PMID: 29119327 DOI: 10.1007/s00441-017-2714-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 10/10/2017] [Indexed: 11/24/2022]
Abstract
In the epididymis, epithelial cells work in a concerted manner to create a luminal environment for sperm maturation, transport, and storage. However, the cell functions may be affected by anthropogenic factors, causing negative impacts on male fertility. In our study, we describe the pattern of protein expression in the epithelium and luminal fluid from epididymis of Oligoryzomys nigripes, a South American sigmodontine rodent whose reproductive biology has been little studied. Nine animals were captured from a preserved area of Atlantic Forest, where the exposure to anthropogenic influences is minimal. Epididymides were processed for histological analysis under light and epifluorescence microscopy, in which we used cell-specific markers aquaporin 9 (AQP9), vacuolar H+-ATPase (V-ATPase), and cytokeratin 5 (KRT5). Other samples were assessed for protein expression using shotgun proteomics. Similar to laboratory rodents, principal cells expressed AQP9 in their stereocilia. Basal cells, identified by KRT5 labeling, presented lateral body projections and a few axiopodia going toward the lumen. Clear cells expressed V-ATPase in their sub-apical vesicles and microplicae, and showed different shapes along the duct. Shotgun proteomics detected 51 proteins from epididymal supernatant. Most of them have been previously described in other species, indicating that they are well conserved. Twenty-three proteins detected in O. nigripes have not been described in epididymis from other South American sigmodontine rodents, confirming that the secretion pattern is species-specific. Our findings in O. nigripes from a protected area may help to create a baseline for studies investigating the effects of anthropogenic factors on functionality of the epididymal epithelium.
Collapse
Affiliation(s)
- Tatiana Prata Menezes
- Department of General Biology, Federal University of Viçosa, Viçosa, MG, 36570-900, Brazil
| | - Eric Hill
- Center for Systems Biology/Program in Membrane Biology/Nephrology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.,Micro Video Instruments, Inc, Avon, MA, 02322, USA
| | | | - Marina D P Lobo
- Laboratory of Proteomics, School of Pharmacy, University of Fortaleza, Ceará, CE, 60811-905, Brazil
| | | | - Sylvie Breton
- Center for Systems Biology/Program in Membrane Biology/Nephrology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Mariana Machado-Neves
- Department of General Biology, Federal University of Viçosa, Viçosa, MG, 36570-900, Brazil.
| |
Collapse
|
21
|
Zhao Z, Xiao S, Yuan X, Yuan J, Zhang C, Li H, Su J, Wang X, Liu Q. AHNAK as a Prognosis Factor Suppresses the Tumor Progression in Glioma. J Cancer 2017; 8:2924-2932. [PMID: 28928883 PMCID: PMC5604443 DOI: 10.7150/jca.20277] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/07/2017] [Indexed: 12/25/2022] Open
Abstract
PURPOSE AHNAK is originally identified as a giant protein based on the estimated size of approximately 700 kDa. The aim of this study is to identify the role of AHNAK in the pathogenesis of glioma. METHODS We tested AHNAK mRNA level in a panel of six human glioma cell lines, and in 30 cases of normal brain tissues and 73 cases of glioma tissue samples using a qRT-PCR method. Further, we analyzed the relationship of AHNAK expression with clinicopathological characteristics in glioma patients. Meanwhile, we analyzed the relationship of expression of AHNAK and survival of glioma patients in survival analyses. Then, in vitro, we analyzed the biological effects of AHNAK in glioma cell lines (U87 and U251) including proliferation assay, cell transwell assay, and apoptosis. And in vivo, we examined the effects of AHNAK on tumor growth using xenograft model of human glioma cells in nude mice. Then we examined the expression of Ki-67-positive cells in these tumors. RESULTS We found that the mRNA levels of AHNAK were down-regulated in 4 of 6 human glioma cell lines, especially in U87 and U251 cell lines. Meanwhile, in glioma patients, a negative correlation was found between the expression of AHNAK and the glioma histopathology. And a low expression of AHNAK was a significant and independent prognostic factor for poor survival of glioma patients. Through over expression of AHNAK in both of U87 and U251, we demonstrated that overexpression of AHNAK could inhibit glioma cell proliferation and invasion, induce apoptosis, and inhibit in vivo glioma tumor growth and ki-67 expression. CONCLUSIONS The AHNAK acts as a potential tumor suppressor. Our study provides a preclinical basis for developing AHNAK as a reliable clinical prognostic indicator for glioma patients, and a new biomarker for treatment response, and a potentially therapeutic target in glioma management options.
Collapse
Affiliation(s)
- Zijin Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Skull Base Surgery and Neuro-oncology at Hunan, Changsha, China
| | - Songhua Xiao
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guanzhou, Guangdong, China
| | - Xianrui Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Skull Base Surgery and Neuro-oncology at Hunan, Changsha, China
| | - Jian Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Skull Base Surgery and Neuro-oncology at Hunan, Changsha, China
| | - Chi Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Skull Base Surgery and Neuro-oncology at Hunan, Changsha, China
| | - Haoyu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Skull Base Surgery and Neuro-oncology at Hunan, Changsha, China
| | - Jun Su
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Skull Base Surgery and Neuro-oncology at Hunan, Changsha, China
| |
Collapse
|
22
|
Chen LH, Chien YW, Liang CT, Chan CH, Fan MH, Huang HY. Green tea extract induces genes related to browning of white adipose tissue and limits weight-gain in high energy diet-fed rat. Food Nutr Res 2017; 61:1347480. [PMID: 28804438 PMCID: PMC5533130 DOI: 10.1080/16546628.2017.1347480] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 06/20/2017] [Indexed: 12/21/2022] Open
Abstract
Background: A wealth of research has reported on the anti-obesity effects of green tea extract (GTE). Although browning of white adipose tissue (WAT) has been reported to attenuate obesity, no study has disclosed the effects of GTE on browning in Sprague Dawley rats. Objectives: The aims of the study were to investigate the effects of GTE on anti-obesity and browning, and their underlying mechanisms. Methods: Four groups of rats (n=10/group) were used including a normal diet with vehicle treatment, and a high-energy diet (HED) with vehicle or GTE by oral gavage at 77.5 or 155 mg/kg/day for 8 weeks. Body weight, fat accumulation, and serum biochemical parameters were used to evaluate obesity. The gene expressions were analyzed using RT-qPCR and western blotting. Results: GTE modulated HED-induced body weight, fat accumulation, and serum levels of triacylglycerol, total cholesterol, low-density lipoprotein, free fatty acids, aspartate aminotransferase, and alanine aminotransferase. Moreover, GTE enhanced the serum high-density lipoprotein. Most importantly, the biomarkers of beige adipose tissue were up-regulated in WAT in GTE-given groups. GTE induced genes involved in different pathways of browning, and reduced transducin-like enhancer protein-3 in WAT. Conclusion: Our results suggest that GTE may improve obesity through inducing browning in HED-fed rats. Abbreviations: ALT: Alanine transaminase; AST: Aspartate transaminase; BAT: Brown adipose tissue; BMP-7: Bone morphogenetic protein-7; BW: Body weight; CIDEA: Cell death activator; CPT-1: Carnitine palmitoyltransferase-1; EFP: Epididymal fat pad; FFA: Free fatty acid; FGF-21: Fibroblast growth factor-21; GTE: Green tea extract; HDL: High-density lipoprotein; HED: high-energy diet; LDL: Low-density lipoprotein; MFP: Mesenteric fat pad; PGC-1α: Activates PPAR-γ coactivator-1; PPAR-γ: Peroxisome proliferator-activated receptor-γ; PRDM-16: PR domain containing 16; RFP: Renal fat pad; SD: Sprague Dawley; TC: Total cholesterol; TG: Triacylglycerol; TLE-3: Transducin-like enhancer protein-3: UCP-1: Uncoupling protein-1; WAT: White adipose tissue.
Collapse
Affiliation(s)
- Li-Han Chen
- YongLin Biomedical Engineering Center, National Taiwan University, Taipei City, Taiwan
| | - Yi-Wen Chien
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei City, Taiwan
| | | | - Ching-Hung Chan
- Department of Food Science, Nutrition, and Nutraceutical Biotechnology, Shih Chien University, Taipei City, Taiwan
| | - Meng-Han Fan
- Department of Food Science, Nutrition, and Nutraceutical Biotechnology, Shih Chien University, Taipei City, Taiwan
| | - Hui-Yu Huang
- Department of Food Science, Nutrition, and Nutraceutical Biotechnology, Shih Chien University, Taipei City, Taiwan
| |
Collapse
|
23
|
Li Y, Liu X, Fan Y, Yang B, Huang C. Radix Stellariae extract prevents high-fat-diet-induced obesity in C57BL/6 mice by accelerating energy metabolism. PeerJ 2017; 5:e3305. [PMID: 28507819 PMCID: PMC5429735 DOI: 10.7717/peerj.3305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/12/2017] [Indexed: 12/22/2022] Open
Abstract
Stellaria dichotoma L. is widely distributed in Ningxia and surrounding areas in northwestern China. Its root, Radix Stellariae (RS), has been used in herbal formulae for treating asthenic-fever, infection, malaria, dyspepsia in children and several other symptoms. This study investigated whether the RS extract (RSE) alleviates metabolic disorders. The results indicated that RSE significantly inhibited body weight gain in high-fat (HF)-diet-fed C57BL/6 mice, reduced fasting glucose levels, and improved insulin tolerance. Moreover, RSE increased the body temperature of the mice and the expression of uncoupling proteins and peroxisome proliferator-activated receptors in the white adipose tissue. Thus, RSE alleviated metabolic disorders in HF-diet-fed C57BL/6 mice by potentially activating UCP and PPAR signaling.
Collapse
Affiliation(s)
- Yin Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Fan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Baican Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|