1
|
Cui L, Li Z, Yang X, Zhou H, Zhang Z, Gao Y, Ren L, Wang Y, Sun R, Ji L, Hua L. Mediating Effect of Insulin-Like Growth Factor-I Underlying the Link Between Vitamin D and Gestational Diabetes Mellitus. Reprod Sci 2024; 31:1541-1550. [PMID: 38347382 DOI: 10.1007/s43032-024-01468-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/19/2024] [Indexed: 05/24/2024]
Abstract
Vitamin D was well-known to be associated with gestational diabetes mellitus (GDM). Insulin-like growth factor-I (IGF-I) has been linked to vitamin D and GDM, respectively. We hypothesize that changes in IGF-I metabolism induced by 25(OH)D3 might contribute to GDM. Therefore, we investigated the independent and combined relationships of serum 25(OH)D3 and IGF-I concentrations with GDM risk, and the mediation effect of IGF-I on 25(OH)D3. A total of 278 pregnant women (including 125 cases and 153 controls) were recruited in our current study. Maternal serum 25(OH)D3 and IGF-I were measured in the second trimester. Logistic regression models were used to estimate the associations of 25(OH)D3 and IGF-I concentrations with the risk of GDM. Mediation analyses were used to explore the mediation effect of IGF-I on the association between 25(OH)D3 and the risk of GDM. After adjusted for the confounded factors, both the third and fourth quartile of 25(OH)D3 decreased the risk of GDM (OR = 0.226; 95% CI, 0.103-0.494; OR = 0.109; 95% CI, 0.045-0.265, respectively) compared to the first quartile of 25(OH)D3. However, the third and fourth quartile of serum IGF-I (OR = 5.174; 95% CI, 2.287-11.705; OR = 12.784; 95% CI, 5.292-30.879, respectively) increased the risk of GDM compared to the first quartile of serum IGF-I. Mediation analyses suggested that 19.62% of the associations between 25(OH)D3 and GDM might be mediated by IGF-I. The lower concentration of serum 25(OH)D3 or higher IGF-I in the second trimester was associated with an increased risk of GDM. The serum IGF-I level might be a potential mediator between 25(OH)D3 and GDM.
Collapse
Affiliation(s)
- Lingling Cui
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Zhiqian Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xiaoli Yang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Huijun Zhou
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Zhengya Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yuting Gao
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Lina Ren
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yibo Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Ruijie Sun
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Linpu Ji
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Linlin Hua
- Department of Advanced Medical Research, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
2
|
Ruhl T, Nuptybayeva A, Kim BS, Beier JP. GPR55 inhibits the pro-adipogenic activity of anandamide in human adipose stromal cells. Exp Cell Res 2024; 435:113908. [PMID: 38163565 DOI: 10.1016/j.yexcr.2023.113908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/22/2023] [Accepted: 12/30/2023] [Indexed: 01/03/2024]
Abstract
The endocannabinoid anandamide (AEA) stimulates adipogenesis via the cannabinoid receptor CB1 in adipose stromal cells (ASCs). However, AEA interacts also with nonclassical cannabinoid receptors, including transient receptor potential cation channel (TRPV)1 and G protein-coupled receptor (GPR)55. Their roles in AEA mediated adipogenesis of human ASCs have not been investigated. We examined the receptor-expressions by immunostaining on human ASCs and tested their functionality by measuring the expression of immediate early genes (IEGs) related to the transcription factor-complex AP-1 upon exposition to receptor agonists. Cells were stimulated with increasing concentrations of specific ligands to investigate the effects on ASC viability (proliferation and metabolic activity), secretory activity, and AEA mediated differentiation. ASCs expressed both receptors, and their activation suppressed IEG expression. TRPV1 did not affect viability or cytokine secretion. GPR55 decreased proliferation, and it inhibited the release of hepatocyte growth factor. Blocking GPR55 increased the pro-adipogenic activity of AEA. These data suggest that GPR55 functions as negative regulator of cannabinoid mediated pro-adipogenic capacity in ASCs.
Collapse
Affiliation(s)
- Tim Ruhl
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany.
| | - Aigul Nuptybayeva
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany.
| | - Bong-Sung Kim
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany; Department of Plastic and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland.
| | - Justus P Beier
- Department of Plastic Surgery, Hand Surgery-Burn Center, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany.
| |
Collapse
|
3
|
Han XX, Zhao FY, Gu KR, Wang GP, Zhang J, Tao R, Yuan J, Gu J, Yu JQ. Development of precocious puberty in children: Surmised medicinal plant treatment. Biomed Pharmacother 2022; 156:113907. [DOI: 10.1016/j.biopha.2022.113907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/29/2022] Open
|
4
|
Lu H, Li ZY, Ding M, Liang C, Weng XQ, Sheng Y, Wu J, Cai X. Trametinib enhances ATRA-induced differentiation in AML cells. Leuk Lymphoma 2021; 62:3361-3372. [PMID: 34355652 DOI: 10.1080/10428194.2021.1961231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
All-trans retinoic acid (ATRA) is only clinically useful in acute promyelocytic leukemia (APL), but not other subtypes of acute myeloid leukemia (AML). In the present study, a clinically achievable concentration of trametinib, a highly selective inhibitor of MEK, enhanced ATRA-induced differentiation in AML cell lines, HL-60 and U937 as well as AML primary cells. Moreover, trametinib-ATRA (tra-ATRA) co-treatment restored ATRA sensitivity in ATRA-resistant AML cell line, HL-60Res. The protein level of STAT3 and the phosphorylation of Akt or JNK were enhanced with tra-ATRA treatment in HL-60, U937, and HL-60Res cells, respectively. Furthermore, tra-ATRA-induced differentiation in HL-60, U937, and HL-60Res cells was inhibited by STAT3, PI3K, and JNK inhibitors, respectively. Therefore, STAT3, Akt, and JNK signaling pathways were involved in tra-ATRA-induced differentiation in HL-60, U937, and HL-60Res cells, respectively. Taken together, our findings may provide novel therapeutic strategies for AML patients.
Collapse
Affiliation(s)
- Hao Lu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ze-Yi Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Ding
- Department of Hematology Oncology, Central Hospital of Minhang District, Shanghai, China
| | - Cui Liang
- Department of Hematology, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xiang-Qin Weng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Sheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xun Cai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Dong K, Zhou WJ, Liu ZH, Hao PJ. The extract of concentrated growth factor enhances osteogenic activity of osteoblast through PI3K/AKT pathway and promotes bone regeneration in vivo. Int J Implant Dent 2021; 7:70. [PMID: 34345951 PMCID: PMC8333229 DOI: 10.1186/s40729-021-00357-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 05/10/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Concentrated growth factor (CGF) is a third-generation platelet concentrate product; the major source of growth factors in CGF is its extract; however, there are few studies on the overall effects of the extract of CGF (CGF-e). The aim of this study was to investigate the effect and mechanism of CGF-e on MC3T3-E1 cells in vitro and to explore the effect of combination of CGF-e and bone collagen (Bio-Oss Collagen, Geistlich, Switzerland) for bone formation in cranial defect model of rats in vivo. METHODS The cell proliferation, ALP activity, mineral deposition, osteogenic-related gene, and protein expression were evaluated in vitro; the newly formed bone was evaluated by histological and immunohistochemical analysis through critical-sized cranial defect rat model in vivo. RESULTS The cell proliferation, ALP activity, mineral deposition, osteogenic-related gene, and protein expression of CGF-e group were significantly increased compared with the control group. In addition, there was significantly more newly formed bone in the CGF-e + bone collagen group, compared to the blank control group and bone collagen only group. CONCLUSIONS CGF-e activated the PI3K/AKT signaling pathway to enhance osteogenic differentiation and mineralization of MC3T3-E1 cells and promoted the bone formation of rat cranial defect model.
Collapse
Affiliation(s)
- Kai Dong
- Department of Dental Implantology, Yantai Stomatological Hospital Affiliated to Binzhou Medical College, No. 142, North Great Str, Yantai, Shandong, 264008, People's Republic of China
| | - Wen-Juan Zhou
- Department of Dental Implantology, Yantai Stomatological Hospital Affiliated to Binzhou Medical College, No. 142, North Great Str, Yantai, Shandong, 264008, People's Republic of China
| | - Zhong-Hao Liu
- Department of Dental Implantology, Yantai Stomatological Hospital Affiliated to Binzhou Medical College, No. 142, North Great Str, Yantai, Shandong, 264008, People's Republic of China
| | - Peng-Jie Hao
- Department of Dental Implantology, Yantai Stomatological Hospital Affiliated to Binzhou Medical College, No. 142, North Great Str, Yantai, Shandong, 264008, People's Republic of China.
| |
Collapse
|
6
|
Tang J, Guo M, Fu J, Ouyang H, Tian Y, Shen X, Huang Y. Polymorphism analysis and expression patterns of the IGF1 gene in the Shitou goose. Arch Anim Breed 2021; 64:315-323. [PMID: 34345653 PMCID: PMC8320522 DOI: 10.5194/aab-64-315-2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 06/15/2021] [Indexed: 11/17/2022] Open
Abstract
Insulin-like growth factor 1 (IGF1) is one of the
endocrine hormones that plays an important role in regulating
growth and development of animals. In this study, polymorphism in the 5′UTR
and 3′UTR coding region and of the IGF1 gene was detected by DNA sequencing
technology, and the abundance of IGF1 mRNA in various tissues at three growth
stages of the Shitou goose was determined by quantitative real-time polymerase chain reaction
(qRT-PCR). Moreover, the differential expression of IGF1 in various tissues
between the Shitou goose and Wuzong goose was revealed. Two single nucleotide
polymorphisms (SNPs) were found in the exon3 region of IGF1 in the Shitou goose. IGF1 mRNA
was extensively expressed in various tissues of Shitou geese with high
abundant expression in the liver, breast muscle and leg muscle at three growth
stages. IGF1 mRNA expression showed a trend of first increase and then decrease
in the pituitary, liver, subcutaneous fat and abdominal fat tissues, but it
decreased in the breast muscle and leg muscle of a Shitou goose with growing age.
Expression of IGF1 in the liver, leg muscle and pituitary tissues of the Shitou goose
was significantly higher than that of the Wuzong goose. This provides a
foundation for further study of regulatory mechanisms of IGF1 in the growth and
development of geese.
Collapse
Affiliation(s)
- Jun Tang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, P.R. China.,Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, Guangdong, 510225, P.R. China
| | - Mao Guo
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, P.R. China
| | - Jing Fu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, P.R. China
| | - Hongjia Ouyang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, P.R. China.,Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, Guangdong, 510225, P.R. China
| | - Yunbo Tian
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, P.R. China.,Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, Guangdong, 510225, P.R. China
| | - Xu Shen
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, P.R. China.,Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, Guangdong, 510225, P.R. China
| | - Yunmao Huang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, P.R. China.,Guangdong Provincial Key Laboratory of Waterfowl Healthy Breeding, Guangzhou, Guangdong, 510225, P.R. China
| |
Collapse
|
7
|
Lu C, Zhao Y, Cao Y, Liu L, Wu S, Li D, Liu S, Xiao S, Wei Y, Li X. MALAT1 Regulated mTOR-Mediated Tau Hyperphosphorylation by Acting as a ceRNA of miR144 in Hippocampus Cells Exposed to High Glucose. Clin Interv Aging 2021; 16:1185-1191. [PMID: 34188461 PMCID: PMC8236260 DOI: 10.2147/cia.s304827] [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: 02/22/2021] [Accepted: 06/03/2021] [Indexed: 11/23/2022] Open
Abstract
Aim High glucose (HG)-induced activation of mTOR promotes tau phosphorylation and leads to diabetes-associated dementia. This study aimed to explore the role of metastasis associated in lung adenocarcinoma transcript 1 (MALAT1) in HG-induced neuronal cell injury. Methods Hippocampus cells were isolated from C57BL/6J mice. After 6 days of culture, the cells were incubated with 5.5 mM glucose in normal medium or 75 mM glucose for 4 days. Cells were transfected with miR-144 mimic, miR-144 inhibitor, siRNA for MALAT1 or corresponding controls. Gene expression was detected by PCR and Western blot analysis. Results HG increased the levels of MALAT1 and p-tau in hippocampal cells. Knockdown of MALAT1 partially reversed the effects of HG on mTOR activity and p-tau protein levels. MALAT1 functioned as competing endogenous RNA (ceRNA) for miR-144, and pre-treatment with MALAT1 siRNA decreased mTOR activity and p-tau protein level in HG-treated hippocampal cells, which was significantly attenuated by miR-144 mimics. Moreover, miR-144 negatively regulated the expression of mTOR and knockdown of MALAT1 suppressed mTOR, while overexpression of mTOR abrogated protective effects of MALAT1 knockdown in HG-treated hippocampal cells. Conclusion MALAT1 knockdown prevented HG-induced mTOR activation and inhibited tau phosphorylation. MALAT1 may be a therapy target for diabetes associated dementia.
Collapse
Affiliation(s)
- Chong Lu
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin, People's Republic of China
| | - Yikui Zhao
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, People's Republic of China
| | - Yan Cao
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Li Liu
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin, People's Republic of China
| | - Shanshan Wu
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin, People's Republic of China
| | - Dongbin Li
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin, People's Republic of China
| | - Shuang Liu
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin, People's Republic of China
| | - Shuyuan Xiao
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin, People's Republic of China
| | - Yafen Wei
- Department of Neurology, Heilongjiang Provincial Hospital, Harbin, People's Republic of China
| | - Xinyu Li
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| |
Collapse
|
8
|
Mukherjee S, Haubner J, Chakraborty A. Targeting the Inositol Pyrophosphate Biosynthetic Enzymes in Metabolic Diseases. Molecules 2020; 25:molecules25061403. [PMID: 32204420 PMCID: PMC7144392 DOI: 10.3390/molecules25061403] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/09/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022] Open
Abstract
In mammals, a family of three inositol hexakisphosphate kinases (IP6Ks) synthesizes the inositol pyrophosphate 5-IP7 from IP6. Genetic deletion of Ip6k1 protects mice from high fat diet induced obesity, insulin resistance and fatty liver. IP6K1 generated 5-IP7 promotes insulin secretion from pancreatic β-cells, whereas it reduces insulin signaling in metabolic tissues by inhibiting the protein kinase Akt. Thus, IP6K1 promotes high fat diet induced hyperinsulinemia and insulin resistance in mice while its deletion has the opposite effects. IP6K1 also promotes fat accumulation in the adipose tissue by inhibiting the protein kinase AMPK mediated energy expenditure. Genetic deletion of Ip6k3 protects mice from age induced fat accumulation and insulin resistance. Accordingly, the pan IP6K inhibitor TNP [N2-(m-trifluorobenzyl), N6-(p-nitrobenzyl)purine] ameliorates obesity, insulin resistance and fatty liver in diet induced obese mice by improving Akt and AMPK mediated insulin sensitivity and energy expenditure. TNP also protects mice from bone loss, myocardial infarction and ischemia reperfusion injury. Thus, the IP6K pathway is a potential target in obesity and other metabolic diseases. Here, we summarize the studies that established IP6Ks as a potential target in metabolic diseases. Further studies will reveal whether inhibition of this pathway has similar pleiotropic benefits on metabolic health of humans.
Collapse
|
9
|
Recovery of muscle mass and muscle oxidative phenotype following disuse does not require GSK-3 inactivation. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165740. [PMID: 32087280 DOI: 10.1016/j.bbadis.2020.165740] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/24/2020] [Accepted: 02/18/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Physical inactivity contributes to muscle wasting and reductions in mitochondrial oxidative phenotype (OXPHEN), reducing physical performance and quality of life during aging and in chronic disease. Previously, it was shown that inactivation of glycogen synthase kinase (GSK)-3β stimulates muscle protein accretion, myogenesis, and mitochondrial biogenesis. Additionally, GSK-3β is inactivated during recovery of disuse-induced muscle atrophy. AIM Therefore, we hypothesize that GSK-3 inhibition is required for reloading-induced recovery of skeletal muscle mass and OXPHEN. METHODS Wild-type (WT) and whole-body constitutively active (C.A.) Ser21/9 GSK-3α/β knock-in mice were subjected to a 14-day hind-limb suspension/14-day reloading protocol. Soleus muscle mass, fiber cross-sectional area (CSA), OXPHEN (abundance of sub-units of oxidative phosphorylation (OXPHOS) complexes and fiber-type composition), as well as expression levels of their main regulators (respectively protein synthesis/degradation, myogenesis and peroxisome proliferator-activated receptor-γ co-activator-1α (PGC-1α) signaling) were monitored. RESULTS Subtle but consistent differences suggesting suppression of protein turnover signaling and decreased expression of several OXPHOS sub-units and PGC-1α signaling constituents were observed at baseline in C.A. GSK-3 versus WT mice. Although soleus mass recovery during reloading occurred more rapidly in C.A. GSK-3 mice, this was not accompanied by a parallel increased CSA. The OXPHEN response to reloading was not distinct between C.A. GSK-3 and WT mice. No consistent or significant differences in reloading-induced changes in the regulatory steps of protein turnover, myogenesis or muscle OXPHEN were observed in C.A. GSK-3 compared to WT muscle. CONCLUSION This study indicates that GSK-3 inactivation is dispensable for reloading-induced recovery of muscle mass and OXPHEN.
Collapse
|
10
|
Vyas KS, Bole M, Vasconez HC, Banuelos JM, Martinez-Jorge J, Tran N, Lemaine V, Mardini S, Bakri K. Profile of Adipose-Derived Stem Cells in Obese and Lean Environments. Aesthetic Plast Surg 2019; 43:1635-1645. [PMID: 31267153 DOI: 10.1007/s00266-019-01397-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 05/04/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND With the demand for stem cells in regenerative medicine, new methods of isolating stem cells are highly sought. Adipose tissue is a readily available and non-controversial source of multipotent stem cells that carries a low risk for potential donors. However, elevated donor body mass index has been associated with an altered cellular microenvironment and thus has implications for stem cell efficacy in recipients. This review explored the literature on adipose-derived stem cells (ASCs) and the effect of donor obesity on cellular function. METHODS A review of published articles on obesity and ASCs was conducted with the PubMed database and the following search terms: obesity, overweight, adipose-derived stem cells and ASCs. Two investigators screened and reviewed the relevant abstracts. RESULTS There is agreement on reduced ASC function in response to obesity in terms of angiogenic differentiation, proliferation, migration, viability, and an altered and inflammatory transcriptome. Osteogenic differentiation and cell yield do not show reasonable agreement. Weight loss partially rescues some of the aforementioned features. CONCLUSIONS Generally, obesity reduces ASC qualities and may have an effect on the therapeutic value of ASCs. Because weight loss and some biomolecules have been shown to rescue these qualities, further research should be conducted on methods to return obese-derived ASCs to baseline. LEVEL V This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors- www.springer.com/00266.
Collapse
Affiliation(s)
- Krishna S Vyas
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA.
| | - Madhav Bole
- Division of Orthopaedic Surgery, London Health Sciences Centre, University Hospital, 339 Windermere Rd., London, ON, N6A 5A5, Canada
| | - Henry C Vasconez
- Division of Plastic Surgery, University of Kentucky, Lexington, KY, USA
| | - Joseph M Banuelos
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Jorys Martinez-Jorge
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Nho Tran
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Valerie Lemaine
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Samir Mardini
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Karim Bakri
- Division of Plastic Surgery, Department of Surgery, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
11
|
Mueed Z, Tandon P, Maurya SK, Deval R, Kamal MA, Poddar NK. Tau and mTOR: The Hotspots for Multifarious Diseases in Alzheimer's Development. Front Neurosci 2019; 12:1017. [PMID: 30686983 PMCID: PMC6335350 DOI: 10.3389/fnins.2018.01017] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 12/17/2018] [Indexed: 12/14/2022] Open
Abstract
The hyperphosphorylation of tau protein and the overexpression of mTOR are considered to be the driving force behind Aβ plaques and Neurofibrillay Tangles (NFT's), hallmarks of Alzheimer's disease (AD). It is now evident that miscellaneous diseases such as Diabetes, Autoimmune diseases, Cancer, etc. are correlated with AD. Therefore, we reviewed the literature on the causes of AD and investigated the association of tau and mTOR with other diseases. We have discussed the role of insulin deficiency in diabetes, activated microglial cells, and dysfunction of blood-brain barrier (BBB) in Autoimmune diseases, Presenilin 1 in skin cancer, increased reactive species in mitochondrial dysfunction and deregulated Cyclins/CDKs in promoting AD pathogenesis. We have also discussed the possible therapeutics for AD such as GSK3 inactivation therapy, Rechaperoning therapy, Immunotherapy, Hormonal therapy, Metal chelators, Cell cycle therapy, γ-secretase modulators, and Cholinesterase and BACE 1-inhibitors which are thought to serve a major role in combating pathological changes coupled with AD. Recent research about the relationship between mTOR and aging and hepatic Aβ degradation offers possible targets to effectively target AD. Future prospects of AD aims at developing novel drugs and modulators that can potentially improve cell to cell signaling, prevent Aβ plaques formation, promote better release of neurotransmitters and prevent hyperphosphorylation of tau.
Collapse
Affiliation(s)
- Zeba Mueed
- Department of Biotechnology, Invertis University, Bareilly, India
| | - Pallavi Tandon
- Department of Biotechnology, Invertis University, Bareilly, India
| | | | - Ravi Deval
- Department of Biotechnology, Invertis University, Bareilly, India
| | - Mohammad A Kamal
- King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Enzymoics, Hebersham, NSW, Australia.,Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | | |
Collapse
|
12
|
Summerfield M, Zhou Y, Zhou T, Wu C, Alpini G, Zhang KK, Xie L. A long-term maternal diet transition from high-fat diet to normal fat diet during pre-pregnancy avoids adipose tissue inflammation in next generation. PLoS One 2018; 13:e0209053. [PMID: 30562363 PMCID: PMC6298692 DOI: 10.1371/journal.pone.0209053] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/27/2018] [Indexed: 12/11/2022] Open
Abstract
Recent studies have suggested that maternal high-fat (HF) diet caused inflammation changes in adipose tissue; however, it remains unclear if maternal diet intervention before pregnancy rescues such effects in offspring. To address this question, female mice were continued on a normal-fat (NF group), or a HF diet (HF group) or transitioned from a HF diet to a NF diet at 1 (H1N group), 5 (H5N group) or 9 weeks (H9N group) prior to pregnancy. Among the three intervention groups, the H9N offspring displayed less and steady body weight gain, and maintained glucose tolerance, whereas the H1N and H5N offspring showed exacerbate these phenotypes. The H1N and H5N, but not the H9N offspring, displayed adipocyte hypertrophy associated with increased expression of genes involved in fat deposition. The H1N and H5N, but not the H9N adipose tissue, displayed increased macrophage infiltration with enhanced expression of inflammatory cytokine genes. In addition, overactivation of the NF-κB and the JNK signaling were observed in the H1N adipose tissue. Overall, our study showed that a long-term but not a short- or medium-term diet intervention before pregnancy released offspring adipose tissue inflammation induced by maternal HF diet, which adds details in our understanding how the maternal environment either promotes or discourages onset of disease in offspring. Clinically, this study is of great value for providing evidence in the design of clinical trials to evaluate the urgently required intervention strategies to minimize the intergenerational cycle of obesity.
Collapse
Affiliation(s)
- Michelle Summerfield
- Department of Nutrition and Food Sciences, Texas A&M University, College Station, TX, United States of America
| | - Yi Zhou
- Department of Nutrition and Food Sciences, Texas A&M University, College Station, TX, United States of America
- Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tianhao Zhou
- Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX, United States of America
| | - Chaodong Wu
- Department of Nutrition and Food Sciences, Texas A&M University, College Station, TX, United States of America
| | - Gianfranco Alpini
- Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX, United States of America
- Research, Central Texas Veterans Health Care System, Temple, TX, United States of America
| | - Ke K. Zhang
- Department of Nutrition and Food Sciences, Texas A&M University, College Station, TX, United States of America
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, College of Medicine, Texas A&M University, Houston, TX, United States of America
| | - Linglin Xie
- Department of Nutrition and Food Sciences, Texas A&M University, College Station, TX, United States of America
- * E-mail:
| |
Collapse
|
13
|
Fernández-Alfonso MS, Somoza B, Tsvetkov D, Kuczmanski A, Dashwood M, Gil-Ortega M. Role of Perivascular Adipose Tissue in Health and Disease. Compr Physiol 2017; 8:23-59. [PMID: 29357124 DOI: 10.1002/cphy.c170004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Perivascular adipose tissue (PVAT) is cushion of fat tissue surrounding blood vessels, which is phenotypically different from other adipose tissue depots. PVAT is composed of adipocytes and stromal vascular fraction, constituted by different populations of immune cells, endothelial cells, and adipose-derived stromal cells. It expresses and releases an important number of vasoactive factors with paracrine effects on vascular structure and function. In healthy individuals, these factors elicit a net anticontractile and anti-inflammatory paracrine effect aimed at meeting hemodynamic and metabolic demands of specific organs and regions of the body. Pathophysiological situations, such as obesity, diabetes or hypertension, induce changes in its amount and in the expression pattern of vasoactive factors leading to a PVAT dysfunction in which the beneficial paracrine influence of PVAT is shifted to a pro-oxidant, proinflammatory, contractile, and trophic environment leading to functional and structural cardiovascular alterations and cardiovascular disease. Many different PVATs surrounding a variety of blood vessels have been described and exhibit regional differences. Both protective and deleterious influence of PVAT differs regionally depending on the specific vascular bed contributing to variations in the susceptibility of arteries and veins to vascular disease. PVAT therefore, might represent a novel target for pharmacological intervention in cardiovascular disease. © 2018 American Physiological Society. Compr Physiol 8:23-59, 2018.
Collapse
Affiliation(s)
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Dmitry Tsvetkov
- Department of Anestesiology, Perioperative and Pain Medicine, HELIOS Klinikum, Berlin-Buch GmbH, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Department of Pharmacology and Experimental Therapy, Eberhard Karls University Hospitals and Clinics, and Interfaculty Center of Pharmacogenomics and Drug Research, Tübingen, Germany
| | - Artur Kuczmanski
- Department of Anestesiology, Perioperative and Pain Medicine, HELIOS Klinikum, Berlin-Buch GmbH, Germany
| | - Mick Dashwood
- Royal Free Hospital Campus, University College Medical School, London, United Kingdom
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| |
Collapse
|
14
|
Maridas DE, DeMambro VE, Le PT, Mohan S, Rosen CJ. IGFBP4 Is Required for Adipogenesis and Influences the Distribution of Adipose Depots. Endocrinology 2017; 158:3488-3500. [PMID: 28938423 PMCID: PMC5659704 DOI: 10.1210/en.2017-00248] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/24/2017] [Indexed: 01/29/2023]
Abstract
Insulinlike growth factor (IGF) I induces adipogenesis in vitro. IGF-binding protein 4 (IGFBP4) is highly expressed in adipocytes and osteoblasts and is inhibitory of IGFs in vitro. We previously reported that Igfbp4 null mice (Igfbp4-/-) had decreased fat proportions at 8 and 16 weeks of age. However, the mechanism leading to the reduced adiposity remains unknown. The purpose of this study was to elucidate how IGFBP4 mediates adipose tissue development in vivo. Our results showed that inguinal and gonadal white adipose tissue (gWAT) from Igfbp4-/- mice had decreased weights and Pparγ expression. Cultures of primary bone marrow stromal cells (BMSCs) and ear mesenchymal stem cells (eMSCs) from mutant mice showed reduced adipogenesis. Both BMSCs and eMSC had a strong induction of Igfbp4 expression during adipogenesis. Furthermore, the increase in phosphorylated Akt (p-Akt), a downstream target of IGF-I signaling, in wild-type cells, was blunted in mutant eMSCs. On a high-fat diet (HFD) there were sexual differences in adipocyte expansion of Igfbp4-/- mice. Mutant males gained weight by expanding their white fat depots. However, Igfbp4-/- female mice were protected against diet-induced obesity. Ovariectomized Igfbp4-/- female mice gained weight in a manner similar to that seen in ovariectomized controls. Thus, Igfbp4 is required for inguinal fat expansion in female mice but not in male mice. However, gWAT expansion, which is prevented by estrogen during HFD, does not require Igfbp4.
Collapse
Affiliation(s)
- David E. Maridas
- Maine Medical Center Research Institute, Scarborough, Maine 04074
| | | | - Phuong T. Le
- Maine Medical Center Research Institute, Scarborough, Maine 04074
| | - Subburaman Mohan
- Veterans Affairs Loma Linda Healthcare System, Loma Linda, California 92357
| | | |
Collapse
|
15
|
Effects of Bariatric Surgery on Change of Brown Adipocyte Tissue and Energy Metabolism in Obese Mice. Obes Surg 2017; 28:820-830. [DOI: 10.1007/s11695-017-2899-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
16
|
Insulin-like growth factor-1 activates different catalytic subunits p110 of PI3K in a cell-type-dependent manner to induce lipogenesis-dependent epithelial–mesenchymal transition through the regulation of ADAM10 and ADAM17. Mol Cell Biochem 2017; 439:199-211. [DOI: 10.1007/s11010-017-3148-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 08/05/2017] [Indexed: 12/23/2022]
|
17
|
Jiang Y, Liu P, Jiao W, Meng J, Feng J. Gax suppresses chemerin/CMKLR1‐induced preadipocyte biofunctions through the inhibition of Akt/mTOR and ERK signaling pathways. J Cell Physiol 2017; 233:572-586. [DOI: 10.1002/jcp.25918] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 03/17/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Yunqi Jiang
- Department of CardiologyThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Ping Liu
- Department of CardiologyThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Wenlin Jiao
- College of PharmacyShandong UniversityJinanShandongChina
| | - Juan Meng
- Department of CardiologyThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Jinbo Feng
- Central LaboratoryThe Qilu Hospital of Shandong UniversityJinanShandongChina
| |
Collapse
|
18
|
Lewitt MS. The Role of the Growth Hormone/Insulin-Like Growth Factor System in Visceral Adiposity. BIOCHEMISTRY INSIGHTS 2017; 10:1178626417703995. [PMID: 28469442 PMCID: PMC5404904 DOI: 10.1177/1178626417703995] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 03/19/2017] [Indexed: 12/18/2022]
Abstract
There is substantial evidence that the growth hormone (GH)/insulin-like growth factor (IGF) system is involved in the pathophysiology of obesity. Both GH and IGF-I have direct effects on adipocyte proliferation and differentiation, and this system is involved in the cross-talk between adipose tissue, liver, and pituitary. Transgenic animal models have been of importance in identifying mechanisms underlying these interactions. It emerges that this system has key roles in visceral adiposity, and there is a rationale for targeting this system in the treatment of visceral obesity associated with GH deficiency, metabolic syndrome, and lipodystrophies. This evidence is reviewed, gaps in knowledge are highlighted, and recommendations are made for future research.
Collapse
Affiliation(s)
- Moira S Lewitt
- School of Health, Nursing & Midwifery, University of the West of Scotland, Paisley, UK
| |
Collapse
|
19
|
Liu J, Fu R, Liu R, Zhao G, Zheng M, Cui H, Li Q, Song J, Wang J, Wen J. Protein Profiles for Muscle Development and Intramuscular Fat Accumulation at Different Post-Hatching Ages in Chickens. PLoS One 2016; 11:e0159722. [PMID: 27508388 PMCID: PMC4980056 DOI: 10.1371/journal.pone.0159722] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/10/2016] [Indexed: 02/06/2023] Open
Abstract
Muscle development and growth influences the efficiency of poultry meat production, and is closely related to deposition of intramuscular fat (IMF), which is crucial in meat quality. To clarify the molecular mechanisms underlying muscle development and IMF deposition in chickens, protein expression profiles were examined in the breast muscle of Beijing-You chickens at ages 1, 56, 98 and 140 days, using isobaric tags for relative and absolute quantification (iTRAQ). Two hundred and four of 494 proteins were expressed differentially. The expression profile at day 1 differed greatly from those at day 56, 98 and 140. KEGG pathway analysis of differential protein expression from pair-wise comparisons (day 1 vs. 56; 56 vs. 98; 98 vs. 140), showed that the fatty acid degradation pathway was more active during the stage from day 1 to 56 than at other periods. This was consistent with the change in IMF content, which was highest at day 1 and declined dramatically thereafter. When muscle growth was most rapid (days 56-98), pathways involved in muscle development were dominant, including hypertrophic cardiomyopathy, dilated cardiomyopathy, cardiac muscle contraction, tight junctions and focal adhesion. In contrast with hatchlings, the fatty acid degradation pathway was downregulated from day 98 to 140, which was consistent with the period for IMF deposition following rapid muscle growth. Changes in some key specific proteins, including fast skeletal muscle troponin T isoform, aldehyde dehydrogenase 1A1 and apolipoprotein A1, were verified by Western blotting, and could be potential biomarkers for IMF deposition in chickens. Protein-protein interaction networks showed that ribosome-related functional modules were clustered in all three stages. However, the functional module involved in the metabolic pathway was only clustered in the first stage (day 1 vs. 56). This study improves our understanding of the molecular mechanisms underlying muscle development and IMF deposition in chickens.
Collapse
Affiliation(s)
- Jie Liu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Ruiqi Fu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Ranran Liu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Guiping Zhao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Maiqing Zheng
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Huanxian Cui
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Qinghe Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Jiao Song
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Jie Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Jie Wen
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| |
Collapse
|
20
|
Katsoulieris EN, Drossopoulou GI, Kotsopoulou ES, Vlahakos DV, Lianos EA, Tsilibary EC. High Glucose Impairs Insulin Signaling in the Glomerulus: An In Vitro and Ex Vivo Approach. PLoS One 2016; 11:e0158873. [PMID: 27434075 PMCID: PMC4951020 DOI: 10.1371/journal.pone.0158873] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/23/2016] [Indexed: 01/14/2023] Open
Abstract
Objective Chronic hyperglycaemia, as seen in type II diabetes, results in both morphological and functional impairments of podocytes in the kidney. We investigated the effects of high glucose (HG) on the insulin signaling pathway, focusing on cell survival and apoptotic markers, in immortalized human glomerular cells (HGEC; podocytes) and isolated glomeruli from healthy rats. Methods and Findings HGEC and isolated glomeruli were cultured for various time intervals under HG concentrations in the presence or absence of insulin. Our findings indicated that exposure of HGEC to HG led to downregulation of all insulin signaling markers tested (IR, p-IR, IRS-1, p-Akt, p-Fox01,03), as well as to increased sensitivity to apoptosis (as seen by increased PARP cleavage, Casp3 activation and DNA fragmentation). Short insulin pulse caused upregulation of insulin signaling markers (IR, p-IR, p-Akt, p-Fox01,03) in a greater extent in normoglycaemic cells compared to hyperglycaemic cells and for the case of p-Akt, in a PI3K-dependent manner. IRS-1 phosphorylation of HG-treated podocytes was negatively regulated, favoring serine versus tyrosine residues. Prolonged insulin treatment caused a significant decrease of IR levels, while alterations in glucose concentrations for various time intervals demonstrated changes of IR, p-IR and p-Akt levels, suggesting that the IR signaling pathway is regulated by glucose levels. Finally, HG exerted similar effects in isolated glomeruli. Conclusions These results suggest that HG compromises the insulin signaling pathway in the glomerulus, promoting a proapoptotic environment, with a possible critical step for this malfunction lying at the level of IRS-1 phosphorylation; thus we herein demonstrate glomerular insulin signaling as another target for investigation for the prevention and/ or treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Elias N. Katsoulieris
- Institute of Biosciences and Applications, National Center for Scientific Research ‘Demokritos’, Athens, Greece
| | - Garyfalia I. Drossopoulou
- Institute of Biosciences and Applications, National Center for Scientific Research ‘Demokritos’, Athens, Greece
- * E-mail: (GID); (ECT)
| | - Eleni S. Kotsopoulou
- Institute of Biosciences and Applications, National Center for Scientific Research ‘Demokritos’, Athens, Greece
| | - Dimitrios V. Vlahakos
- 2nd Department of Propaedeutic Medicine, Attikon University Hospital, Athens, Greece
| | - Elias A. Lianos
- Department of Pathology, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Effie C. Tsilibary
- Institute of Biosciences and Applications, National Center for Scientific Research ‘Demokritos’, Athens, Greece
- * E-mail: (GID); (ECT)
| |
Collapse
|
21
|
Tharp WG, Gupta D, Smith J, Jones KP, Jones AM, Pratley RE. Effects of glucose and insulin on secretion of amyloid-β by human adipose tissue cells. Obesity (Silver Spring) 2016; 24:1471-9. [PMID: 27172814 PMCID: PMC5084817 DOI: 10.1002/oby.21494] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/05/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Obesity and type 2 diabetes mellitus are risk factors for developing Alzheimer disease. Overlapping patterns of metabolic dysfunction may be common molecular links between these complex diseases. Amyloid-β (Aβ) precursor protein and associated β- and γ-secretases are expressed in adipose tissue. Aβ precursor protein is up-regulated with obesity and correlated to insulin resistance. Aβ may be secreted by adipose tissue, its production may be regulated through metabolic pathways, and Aβ may exert effects on adipose tissue insulin receptor signaling. METHODS Human stromal-vascular cells and differentiated adipocytes were cultured with different combinations of glucose and insulin and then assayed for Aβ in conditioned media. Aβ was measured in vivo using adipose tissue microdialysis. RESULTS Aβ secretion was increased by glucose and insulin in vitro. Adipose tissue microdialysates contained Aβ. Adipocytes treated with Aβ had decreased expression of insulin receptor substrate-2 and reduced Akt-1 phosphorylation. CONCLUSIONS Aβ was made by adipose tissue cells in vitro at concentrations similar to in vivo measurements. Regulation of Aβ production by glucose and insulin and effects of Aβ on the insulin receptor pathway suggest similar cellular mechanisms may exist between neuronal dysfunction in Alzheimer disease and adipose dysfunction in type 2 diabetes.
Collapse
Affiliation(s)
- William G. Tharp
- Department of Medicine Endocrine UnitUniversity of Vermont College of MedicineBurlingtonVermontUSA
| | - Dhananjay Gupta
- Department of Medicine Endocrine UnitUniversity of Vermont College of MedicineBurlingtonVermontUSA
| | - Joshua Smith
- Florida Hospital Sanford/Burnham Translational Research Institute for Metabolism and DiabetesOrlandoFloridaUSA
| | - Karen P. Jones
- Florida Hospital Sanford/Burnham Translational Research Institute for Metabolism and DiabetesOrlandoFloridaUSA
| | - Amanda M. Jones
- Florida Hospital Sanford/Burnham Translational Research Institute for Metabolism and DiabetesOrlandoFloridaUSA
| | - Richard E. Pratley
- Florida Hospital Sanford/Burnham Translational Research Institute for Metabolism and DiabetesOrlandoFloridaUSA
| |
Collapse
|
22
|
Does Metabolically Healthy Obesity Exist? Nutrients 2016; 8:nu8060320. [PMID: 27258304 PMCID: PMC4924161 DOI: 10.3390/nu8060320] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/04/2016] [Accepted: 05/17/2016] [Indexed: 02/07/2023] Open
Abstract
The relationship between obesity and other metabolic diseases have been deeply studied. However, there are clinical inconsistencies, exceptions to the paradigm of "more fat means more metabolic disease", and the subjects in this condition are referred to as metabolically healthy obese (MHO).They have long-standing obesity and morbid obesity but can be considered healthy despite their high degree of obesity. We describe the variable definitions of MHO, the underlying mechanisms that can explain the existence of this phenotype caused by greater adipose tissue inflammation or the different capacity for adipose tissue expansion and functionality apart from other unknown mechanisms. We analyze whether these subjects improve after an intervention (traditional lifestyle recommendations or bariatric surgery) or if they stay healthy as the years pass. MHO is common among the obese population and constitutes a unique subset of characteristics that reduce metabolic and cardiovascular risk factors despite the presence of excessive fat mass. The protective factors that grant a healthier profile to individuals with MHO are being elucidated.
Collapse
|
23
|
Kim Y, Kim C, Son SM, Song H, Hong HS, Han SH, Mook-Jung I. The novel RAGE interactor PRAK is associated with autophagy signaling in Alzheimer's disease pathogenesis. Mol Neurodegener 2016; 11:4. [PMID: 26758977 PMCID: PMC4709948 DOI: 10.1186/s13024-016-0068-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 01/04/2016] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The receptor for advanced glycation end products (RAGE) has been found to interact with amyloid β (Aβ). Although RAGE does not have any kinase motifs in its cytosolic domain, the interaction between RAGE and Aβ triggers multiple cellular signaling involved in Alzheimer's disease (AD). However, the mechanism of signal transduction by RAGE remains still unknown. Therefore, identifying binding proteins of RAGE may provide novel therapeutic targets for AD. RESULTS In this study, we identified p38-regulated/activated protein kinase (PRAK) as a novel RAGE interacting molecule. To investigate the effect of Aβ on PRAK mediated RAGE signaling pathway, we treated SH-SY5Y cells with monomeric form of Aβ. We demonstrated that Aβ significantly increased the phosphorylation of PRAK as well as the interaction between PRAK and RAGE. We showed that knockdown of PRAK rescued mTORC1 inactivation induced by Aβ treatment and decreased the formation of Aβ-induced autophagosome. CONCLUSIONS We provide evidence that PRAK plays a critical role in AD pathology as a key interactor of RAGE. Thus, our data suggest that PRAK might be a potential therapeutic target of AD involved in RAGE-mediated cell signaling induced by Aβ.
Collapse
Affiliation(s)
- Yoonhee Kim
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Chaeyoung Kim
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Sung Min Son
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Hyundong Song
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Hyun Seok Hong
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Sun-ho Han
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| |
Collapse
|
24
|
Zhang Y, Nowicka A, Solley TN, Wei C, Parikh A, Court L, Burks JK, Andreeff M, Woodward WA, Dadbin A, Kolonin MG, Lu KH, Klopp AH. Stromal Cells Derived from Visceral and Obese Adipose Tissue Promote Growth of Ovarian Cancers. PLoS One 2015; 10:e0136361. [PMID: 26317219 PMCID: PMC4552684 DOI: 10.1371/journal.pone.0136361] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/31/2015] [Indexed: 01/23/2023] Open
Abstract
Obesity, and in particular visceral obesity, has been associated with an increased risk of developing cancers as well as higher rates of mortality following diagnosis. The impact of obesity on adipose-derived stromal cells (ASC), which contribute to the formation of tumor stroma, is unknown. Here we hypothesized that visceral source and diet-induced obesity (DIO) changes the ASC phenotype, contributing to the tumor promoting effects of obesity. We found that ASC isolated from subcutaneous (SC-ASC) and visceral (V-ASC) white adipose tissue(WAT) of lean(Le) and obese(Ob) mice exhibited similar mesenchymal cell surface markers expression, and had comparable effects on ovarian cancer cell proliferation and migration. Obese and visceral derived ASC proliferated slower and exhibited impaired differentiation into adipocytes and osteocytes in vitro as compared to ASC derived from subcutaneous WAT of lean mice. Intraperitoneal co-injection of ovarian cancer cells with obese or visceral derived ASC, but not lean SC-ASC, increased growth of intraperitoneal ID8 tumors as compared to controls. Obese and V-ASC increased stromal infiltration of inflammatory cells, including CD3+ T cells and F4/80+ macrophages. Obese and visceral derived ASC, but not lean SC-ASC, increased expression of chemotactic factors IL-6, MIP-2, and MCP-1 when cultured with tumor cells. Overall, these results demonstrate that obese and V-ASC have a unique phenotype, with more limited proliferation and differentiation capacity but enhanced expression of chemotactic factors in response to malignant cells which support infiltration of inflammatory cells and support tumor growth and dissemination.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Aleksandra Nowicka
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Travis N. Solley
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Caimiao Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Aaroh Parikh
- Department of Physics and Astronomy, Rice University, Houston, Texas, United States of America
| | - Laurence Court
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Jared K. Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Wendy A. Woodward
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Ali Dadbin
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Karen H. Lu
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Ann H. Klopp
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
25
|
Liu P, Kong F, Wang J, Lu Q, Xu H, Qi T, Meng J. Involvement of IGF-1 and MEOX2 in PI3K/Akt1/2 and ERK1/2 pathways mediated proliferation and differentiation of perivascular adipocytes. Exp Cell Res 2015; 331:82-96. [DOI: 10.1016/j.yexcr.2014.09.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/02/2014] [Accepted: 09/07/2014] [Indexed: 01/20/2023]
|
26
|
Abstract
Alzheimer's disease (AD) is characterized by cognitive impairment in clinical presentation, and by β-amyloid (Aβ) production and the hyper-phosphorylation of tau in basic research. More highlights demonstrate that the activation of the mammalian target of rapamycin (mTOR) enhances Aβ generation and deposition by modulating amyloid precursor protein (APP) metabolism and upregulating β- and γ-secretases. mTOR, an inhibitor of autophagy, decreases Aβ clearance by scissoring autophagy function. mTOR regulates Aβ generation or Aβ clearance by regulating several key signaling pathways, including phosphoinositide 3-kinase (PI3-K)/protein kinase B (Akt), glycogen synthase kinase 3 [GSK-3], AMP-activated protein kinase (AMPK), and insulin/insulin-like growth factor 1 (IGF-1). The activation of mTOR is also a contributor to aberrant hyperphosphorylated tau. Rapamycin, the inhibitor of mTOR, may mitigate cognitive impairment and inhibit the pathologies associated with amyloid plaques and neurofibrillary tangles by promoting autophagy. Furthermore, the upstream and downstream components of mTOR signaling are involved in the pathogenesis and progression of AD. Hence, inhibiting the activation of mTOR may be an important therapeutic target for AD.
Collapse
Affiliation(s)
- Zhiyou Cai
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan Renmin Hospital, Shiyan, Hubei Province, People's Republic of China
| | - Guanghui Chen
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan Renmin Hospital, Shiyan, Hubei Province, People's Republic of China
| | - Wenbo He
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan Renmin Hospital, Shiyan, Hubei Province, People's Republic of China
| | - Ming Xiao
- Department of Anatomy, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Liang-Jun Yan
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
27
|
Moreno-Indias I, Tinahones FJ. Impaired adipose tissue expandability and lipogenic capacities as ones of the main causes of metabolic disorders. J Diabetes Res 2015; 2015:970375. [PMID: 25922847 PMCID: PMC4398959 DOI: 10.1155/2015/970375] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 03/15/2015] [Accepted: 03/18/2015] [Indexed: 02/07/2023] Open
Abstract
Obesity is considered a major health problem. However, mechanisms involved and its comorbidities are not elucidated. Recent theories concerning the causes of obesity have focused on a limit to the functional capacity of adipose tissue, comparing it with other vital organs. This assumption has been the central point of interest in our laboratory. We proposed that the failure of adipose tissue is initiated by the difficulty of this tissue to increase its cellularity due to excess in fat contribution, owing to genetic or environmental factors. Nevertheless, why the adipose tissue reduces its capacity to make new adipocytes via mesenchymal cells of the stroma has not yet been elucidated. Thus, we suggest that this tissue ceases fulfilling its main function, the storage of excess fat, thereby affecting some of the key factors involved in lipogenesis, some of which are reviewed in this paper (PPARγ, ROR1, FASN, SCD1, Rab18, BrCa1, ZAG, and FABP4). On the other hand, mechanisms involved in adipose tissue expandability are also impaired, predominating hypertrophy via an increase in apoptosis and a decrease in adipogenesis and angiogenesis. However, adipose tissue failure is only part of this great orchestra, only a chapter of this nightmare.
Collapse
Affiliation(s)
- Isabel Moreno-Indias
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Complejo Hospitalario de Málaga (Virgen de la Victoria), Universidad de Málaga, 29010 Málaga, Spain
- Ciber Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- *Isabel Moreno-Indias: and
| | - Francisco José Tinahones
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Complejo Hospitalario de Málaga (Virgen de la Victoria), Universidad de Málaga, 29010 Málaga, Spain
- Ciber Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
- *Francisco José Tinahones:
| |
Collapse
|
28
|
Liu P, Feng J, Kong F, Lu Q, Xu H, Meng J, Jiang Y. Gax inhibits perivascular preadipocyte biofunction mediated by IGF-1 induced FAK/Pyk2 and ERK2 cooperative pathways. Cell Signal 2014; 26:3036-45. [DOI: 10.1016/j.cellsig.2014.09.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/25/2014] [Indexed: 02/06/2023]
|
29
|
Viesti A. Collares R, Salgado W, Pretti da Cunha Tirapelli D, dos Santos JS. The expression of LEP, LEPR, IGF1 and IL10 in obesity and the relationship with microRNAs. PLoS One 2014; 9:e93512. [PMID: 24690978 PMCID: PMC3972109 DOI: 10.1371/journal.pone.0093512] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 03/07/2014] [Indexed: 11/19/2022] Open
Abstract
Obesity is a multifactorial disease, with epigenetic alterations. Have been described modifications in the expression of some microRNAs, and some proteins related to obesity. The objective was to determine and correlate, in obese patients, the gene expression of LEP, LEPR, IGF1, IL10 and of miR-27a, miR-27b, miR-143 and miR-145. RNA was extracted from biopsies of subcutaneous fat, liver and visceral fat of 15 obese subjects submitted to bariatric surgery and of 15 non-obese subjects submitted to cholecystectomy for cDNA synthesis and for RT-PCR. The microRNAs were chosen using the TargetScan software. An increased expression of LEP and IGF1 was detected in the subcutaneous fat of the obese group compared to control, while the expression of IGF1 was higher in the control group than in the obese one. MiRNA-27a had a higher expression in the omentum of the obese patients and there was also a correlation in the expression of miRNA-145 and LEPR in the omentum of this group.
Collapse
Affiliation(s)
- Renata Viesti A. Collares
- Department of Surgery and Anatomy of the Faculty of Medicine of Ribeirao Preto, University of São Paulo, Ribeirao Preto, SP, Brazil
| | - Wilson Salgado
- Department of Surgery and Anatomy of the Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirao Preto, SP, Brazil
- * E-mail:
| | - Daniela Pretti da Cunha Tirapelli
- Department of Surgery and Anatomy of the Faculty of Medicine of Ribeirão Preto, University of São Paulo and Laboratory of Molecular Biology, Ribeirao Preto, SP, Brazil
| | - José Sebastião dos Santos
- Department of Surgery and Anatomy of the Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirao Preto, SP, Brazil
| |
Collapse
|
30
|
Conover CA, Harstad SL, Tchkonia T, Kirkland JL. Preferential impact of pregnancy-associated plasma protein-A deficiency on visceral fat in mice on high-fat diet. Am J Physiol Endocrinol Metab 2013; 305:E1145-53. [PMID: 24045868 PMCID: PMC3840208 DOI: 10.1152/ajpendo.00405.2013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Accumulation of visceral fat, more so than subcutaneous fat, is strongly associated with severe metabolic complications. However, the factors regulating depot-specific adipogenesis are poorly understood. In this study, we show differential expression of pregnancy-associated plasma protein-A (PAPP-A), a secreted regulator of local insulin-like growth factor (IGF) action, in adipose tissue of mice. PAPP-A mRNA expression was fivefold higher in visceral (mesenteric) fat compared with subcutaneous (inguinal, subscapular), perirenal, and brown fat of mice. To investigate the possible role of depot-specific PAPP-A expression in fat accumulation, wild-type (WT) and PAPP-A knockout (KO) mice were fed a high-fat diet (HFD) for up to 20 wk. Adipocyte size increased in subcutaneous and perirenal depots similarly in WT and PAPP-A KO mice. However, fat cell size and in vivo lipid uptake were significantly reduced in mesenteric fat of PAPP-A KO compared with WT mice. After 20 wk on HFD, phosphorylation of AKT, a downstream signaling intermediate of IGF-I and insulin receptor activation, was significantly decreased by 50% in mesenteric compared with subcutaneous fat in WT mice, but was significantly increased threefold in mesenteric compared with subcutaneous fat in PAPP-A KO mice. This appeared to be because of enhanced insulin-stimulated signaling in mesenteric fat of PAPP-A KO mice. These data establish fat depot-specific expression of PAPP-A and indicate preferential impact of PAPP-A deficiency on visceral fat in the mouse that is associated with enhanced insulin receptor signaling. Thus, PAPP-A may be a potential target for treatment and/or prevention strategies for visceral obesity and related morbidities.
Collapse
Affiliation(s)
- Cheryl A Conover
- Division of Endocrinology, Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota; and
| | | | | | | |
Collapse
|
31
|
Dünner N, Quezada C, Berndt FA, Cánovas J, Rojas CV. Angiotensin II signaling in human preadipose cells: participation of ERK1,2-dependent modulation of Akt. PLoS One 2013; 8:e75440. [PMID: 24098385 PMCID: PMC3788799 DOI: 10.1371/journal.pone.0075440] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 08/14/2013] [Indexed: 12/13/2022] Open
Abstract
The renin-angiotensin system expressed in adipose tissue has been implicated in the modulation of adipocyte formation, glucose metabolism, triglyceride accumulation, lipolysis, and the onset of the adverse metabolic consequences of obesity. As we investigated angiotensin II signal transduction mechanisms in human preadipose cells, an interplay of extracellular-signal-regulated kinases 1 and 2 (ERK1,2) and Akt/PKB became evident. Angiotensin II caused attenuation of phosphorylated Akt (p-Akt), at serine 473; the p-Akt/Akt ratio decreased to 0.5±0.2-fold the control value without angiotensin II (p<0.001). Here we report that the reduction of phosphorylated Akt associates with ERK1,2 activities. In the absence of angiotensin II, inhibition of ERK1,2 activation with U0126 or PD98059 resulted in a 2.1±0.5 (p<0.001) and 1.4±0.2-fold (p<0.05) increase in the p-Akt/Akt ratio, respectively. In addition, partial knockdown of ERK1 protein expression by the short hairpin RNA technique also raised phosphorylated Akt in these cells (the p-Akt/Akt ratio was 1.5±0.1-fold the corresponding control; p<0.05). Furthermore, inhibition of ERK1,2 activation with U0126 prevented the reduction of p-Akt/Akt by angiotensin II. An analogous effect was found on the phosphorylation status of Akt downstream effectors, the forkhead box (Fox) proteins O1 and O4. Altogether, these results indicate that angiotensin II signaling in human preadipose cells involves an ERK1,2-dependent attenuation of Akt activity, whose impact on the biological functions under its regulation is not fully understood.
Collapse
Affiliation(s)
- Natalia Dünner
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Carolina Quezada
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - F. Andrés Berndt
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - José Cánovas
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Cecilia V. Rojas
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Institute of Nutrition and Food Technology, Universidad de Chile, Santiago, Chile
- * E-mail:
| |
Collapse
|
32
|
El-Mesallamy HO, Hamdy NM, Sallam AAM. Effect of obesity and glycemic control on serum lipocalins and insulin-like growth factor axis in type 2 diabetic patients. Acta Diabetol 2013; 50:679-85. [PMID: 22307870 DOI: 10.1007/s00592-012-0373-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 01/16/2012] [Indexed: 01/15/2023]
Abstract
Obesity, insulin resistance (IR), and progressive decline in pancreatic β-cell function are major features of type 2 diabetes mellitus (T2DM). Altered adipokines contribute to obesity-induced IR. Hence understanding of adipokines' relation to obesity and glycemic control could be useful to improve disease outcomes. We aimed at determination of serum retinol binding protein-4 (RBP-4), lipocalin-2 (LCN-2), insulin-like growth factor-I (IGF-I), and its binding protein-3 (IGFBP-3) levels in T2DM patients with the impact of obesity and glycemic control on them and their relation to β-cell function. Serum insulin, RBP-4, LCN-2, IGF-I, and IGFBP-3 estimated by ELISA were examined in 32 T2DM patients and age- and sex-matched 20 healthy controls. Significant elevation was observed in serum RBP-4 (P < 0.001), LCN-2 (P < 0.01), and IGF-I/IGFBP-3 molar ratio (P < 0.05) in T2DM patients in comparison with healthy controls. There was no significant difference in them between nonobese and obese diabetics. However, RBP-4 and IGF/IGFBP-3 molar ratio were higher in uncontrolled than in controlled diabetic patients at P < 0.001 and P < 0.01, respectively. Moreover, RBP-4, LCN-2, and IGF-I/IGFBP-3 molar ratio were negatively correlated with β-cell function. In conclusion, serum RBP-4 and IGF-I/IGFBP-3 molar ratio but not LCN-2 were prominently elevated with poor glycemic control rather than obesity in T2DM patients. Whereas, declining β-cell function is associated with elevation of serum RBP-4, LCN-2 as well as IGF-I/IGFBP-3 molar ratio.
Collapse
Affiliation(s)
- Hala O El-Mesallamy
- Biochemistry Department, Faculty of Pharmacy, Ain-Shams University, Abassia, Cairo, 11566, Egypt
| | | | | |
Collapse
|
33
|
Neacsu O, Cleveland K, Xu H, Tchkonia TT, Kirkland JL, Boney CM. IGF-I attenuates FFA-induced activation of JNK1 phosphorylation and TNFα expression in human subcutaneous preadipocytes. Obesity (Silver Spring) 2013; 21:1843-9. [PMID: 23512893 PMCID: PMC3690156 DOI: 10.1002/oby.20329] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 12/12/2012] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Free fatty acids (FFAs) are increased in visceral fat and contribute to insulin resistance through multiple mechanisms, including c-Jun N-terminal kinase (JNK) activation and expression of TNFα. Given that insulin-like growth factor-1 (IGF-1)-mediated proliferation is impaired in omental compared to subcutaneous (SC) preadipocytes, we investigated IGF-I anti-inflammatory action in preadipocytes from SC and omental adipose tissue. DESIGN AND METHODS Preadipocytes isolated from abdominal SC and omental fat of obese subjects were studied in primary culture. Cells were exposed to FFAs with or without IGF-I pretreatment followed by analysis of cytokine expression and JNK phosphorylation. Lentivirus infection was used to express a constitutively active AKT (myr-AKT) in omental preadipocytes. RESULTS FFAs increased the expression of tumor necrosis factor (TNF)α, interleukin (IL)-6, and monocyte chemotactic protein (MCP)-1 in SC and omental preadipocytes. IGF-I pretreatment reduced FFA-induced JNK1 phosphorylation and TNFα expression in SC but not omental preadipocytes. Treatment with the JNK1/2 inhibitor SP600125 reduced FFA-induced expression of TNFα. FFAs and MALP-2, a specific TLR2/6 ligand, but not specific ligands for TLR4 and TLR1/2, increased JNK1 phosphorylation. IGF-I completely inhibited MALP-2-stimulated phosphorylation of JNK1. Expression of myr-AKT in omental preadipocytes inhibited FFA-stimulated JNK1 phosphorylation. CONCLUSIONS IGF-I attenuated FFA-induced JNK1 phosphorylation and TNFα expression through activation of AKT in human subcutaneous but not omental preadipocytes.
Collapse
Affiliation(s)
- Otilia Neacsu
- Department of Pediatrics, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence RI, 02903
| | - Kelly Cleveland
- Department of Pediatrics, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence RI, 02903
| | - Haiyan Xu
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence RI, 02903
| | - Tamara T. Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester MN, 55905
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester MN, 55905
| | - Charlotte M Boney
- Department of Pediatrics, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence RI, 02903
- corresponding author/reprint requests: Charlotte M Boney MD, Department of Pediatrics, Rhode Island Hospital and Brown University, 593 Eddy St, MPS-2, Providence RI 02903, 401-444-7891 phone, 401-444-2534 fax,
| |
Collapse
|
34
|
Tchkonia T, Thomou T, Zhu Y, Karagiannides I, Pothoulakis C, Jensen MD, Kirkland JL. Mechanisms and metabolic implications of regional differences among fat depots. Cell Metab 2013; 17:644-656. [PMID: 23583168 PMCID: PMC3942783 DOI: 10.1016/j.cmet.2013.03.008] [Citation(s) in RCA: 492] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fat distribution is closely linked to metabolic disease risk. Distribution varies with sex, genetic background, disease state, certain drugs and hormones, development, and aging. Preadipocyte replication and differentiation, developmental gene expression, susceptibility to apoptosis and cellular senescence, vascularity, inflammatory cell infiltration, and adipokine secretion vary among depots, as do fatty-acid handling and mechanisms of enlargement with positive-energy and loss with negative-energy balance. How interdepot differences in these molecular, cellular, and pathophysiological properties are related is incompletely understood. Whether fat redistribution causes metabolic disease or whether it is a marker of underlying processes that are primarily responsible is an open question.
Collapse
Affiliation(s)
| | - Thomas Thomou
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging
| | - Iordanes Karagiannides
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, Department of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Charalabos Pothoulakis
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, Department of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
35
|
Czech MP, Tencerova M, Pedersen DJ, Aouadi M. Insulin signalling mechanisms for triacylglycerol storage. Diabetologia 2013; 56:949-64. [PMID: 23443243 PMCID: PMC3652374 DOI: 10.1007/s00125-013-2869-1] [Citation(s) in RCA: 178] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 01/22/2013] [Indexed: 02/06/2023]
Abstract
Insulin signalling is uniquely required for storing energy as fat in humans. While de novo synthesis of fatty acids and triacylglycerol occurs mostly in liver, adipose tissue is the primary site for triacylglycerol storage. Insulin signalling mechanisms in adipose tissue that stimulate hydrolysis of circulating triacylglycerol, uptake of the released fatty acids and their conversion to triacylglycerol are poorly understood. New findings include (1) activation of DNA-dependent protein kinase to stimulate upstream stimulatory factor (USF)1/USF2 heterodimers, enhancing the lipogenic transcription factor sterol regulatory element binding protein 1c (SREBP1c); (2) stimulation of fatty acid synthase through AMP kinase modulation; (3) mobilisation of lipid droplet proteins to promote retention of triacylglycerol; and (4) upregulation of a novel carbohydrate response element binding protein β isoform that potently stimulates transcription of lipogenic enzymes. Additionally, insulin signalling through mammalian target of rapamycin to activate transcription and processing of SREBP1c described in liver may apply to adipose tissue. Paradoxically, insulin resistance in obesity and type 2 diabetes is associated with increased triacylglycerol synthesis in liver, while it is decreased in adipose tissue. This and other mysteries about insulin signalling and insulin resistance in adipose tissue make this topic especially fertile for future research.
Collapse
Affiliation(s)
- M P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, MA 01605, USA.
| | | | | | | |
Collapse
|
36
|
Kamycheva E, Berg V, Jorde R. Insulin-like growth factor I, growth hormone, and insulin sensitivity: the effects of a one-year cholecalciferol supplementation in middle-aged overweight and obese subjects. Endocrine 2013; 43:412-8. [PMID: 23109222 DOI: 10.1007/s12020-012-9825-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 10/19/2012] [Indexed: 12/24/2022]
Abstract
Both altered GH-IGF-I axis and low serum levels of 25-hydroxyvitamin D (25(OH)D) are linked to measures of metabolic syndrome. Our hypothesis was that there is a relation between GH, IGF-I, and 25(OH)D; and that vitamin D supplementation may have an effect on the levels of GH, IGF-I, and IGF-I/IGFBP-3 ratio. 318 overweight and obese subjects completed a one-year randomized intervention with either 40,000 or 20,000 IU cholecalciferol per week or placebo. GH, IGF-I, IGFBP-3 and measures of insulin resistance were evaluated at baseline and at the end of study. There was a significant relation between entities of GH-IGF-I axis and insulin resistance. Subjects with severe obesity had significantly lower serum 25(OH)D and had a significant linear decline in IGF-I/IGFBP-3 ratio with increasing serum 25(OH)D quartiles. Vitamin D status was an independent predictor of GH-IGF-I axis and supplementation with vitamin D decreased IGF-I/IGFBP-3 ratio in subjects without severe obesity. No corresponding effect of vitamin D supplementation on BMI or insulin resistance was observed. Adverse effects of GH-IGF-I axis on glucose metabolism and the development of metabolic syndrome may be in part associated with the changes in vitamin D status.
Collapse
Affiliation(s)
- Elena Kamycheva
- Department of Medicine, University Hospital of North Norway, 9038, Tromsø, Norway.
| | | | | |
Collapse
|
37
|
Zhao P, Deng Y, Gu P, Wang Y, Zhou H, Hu Y, Chen P, Fan X. Insulin-like growth factor 1 promotes the proliferation and adipogenesis of orbital adipose-derived stromal cells in thyroid-associated ophthalmopathy. Exp Eye Res 2012; 107:65-73. [PMID: 23219871 DOI: 10.1016/j.exer.2012.11.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 11/23/2012] [Accepted: 11/26/2012] [Indexed: 12/20/2022]
Abstract
Thyroid-associated ophthalmopathy (TAO) is characterised by increased volume of the orbital contents involving adipose tissue, but the factors responsible for stimulation of orbital adipogenesis remain uncertain. Previous studies have shown that insulin-like growth factor 1 (IGF-1) is increased in the orbital fatty connective tissues of patients with TAO. The present study was conducted to investigate the effects of IGF-1 on orbital adipose-derived stromal cells (OADSCs) derived from TAO patients and to identify the signalling mechanisms involved. Our results showed that IGF-1 significantly promoted the cell proliferation and lipid accumulation of TAO OADSCs. The mRNA expression of adipogenic markers (adiponectin, leptin, adipocyte fatty acid binding protein [AP2] and fatty acid synthase [FAS]) was increased in TAO cultures treated with IGF-1. Further research demonstrated that the protein levels of peroxisome proliferator-activated receptor-γ (PPARγ) were up-regulated when OADSCs were treated with IGF-1. We also found that the inhibition of either IGF-1 receptor (IGF-1R) or phosphoinositide 3-kinase (PI3K) activity decreased the levels of IGF-1-stimulated mRNA encoding adiponectin, leptin, AP2, and FAS, as well as PPARγ protein levels. Moreover, the expression of phosphorylated Akt (p-Akt) protein in TAO cells was up-regulated by IGF-1, while a specific PI3K inhibitor (LY294002) or an antibody of IGF-1R blocked this effect. These results indicate that IGF-1 is a pro-proliferative and pro-adipogenic factor in TAO OADSCs. IGF-1 enhances the adipogenesis of TAO OADSCs by up-regulation of PPARγ via the activation of the IGF-1R and PI3K pathways, suggesting that the blocking of IGF-1R or inhibition of PI3K signalling might be a potential novel therapeutic approach to TAO.
Collapse
Affiliation(s)
- Pingqian Zhao
- Department of Ophthalmology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhizaoju Road, Shanghai 200011, China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Garten A, Schuster S, Kiess W. The insulin-like growth factors in adipogenesis and obesity. Endocrinol Metab Clin North Am 2012; 41:283-95, v-vi. [PMID: 22682631 DOI: 10.1016/j.ecl.2012.04.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adipose tissue has been recognized as a major target of growth hormone (GH) action. GH was shown to inhibit adipocyte differentiation but stimulated preadipocyte proliferation in vitro. GH acts directly via its receptor or via upregulating insulin-like growth factor (IGF)-I, which is a critical mediator of preadipocyte proliferation, differentiation, and survival. Results from clinical studies on GH treatment in patients with GH deficiency or GH insensitivity syndrome can be used to dissect GH and IGF as well as IGF-binding protein (IGFBP) actions in vivo. In this article, changes of the GH/IGF system during adipocyte differentiation in vitro as well as related signaling pathways and their impact on adipose tissue growth and function are discussed. Clinical considerations include the effects of GH and IGF-I on adipose tissue during treatment of GH deficiency, differences in the IGF system between visceral and subcutaneous adipose tissue depots as well as the recently emerging role for adipose tissue in the regulation of glucose homeostasis.
Collapse
Affiliation(s)
- A Garten
- Department of Women and Child Health, Hospital for Children and Adolescents, Center for Pediatric Research Leipzig, University Hospitals, Liebigstraße 20a, 04103 Leipzig, Germany
| | | | | |
Collapse
|
39
|
Baglioni S, Cantini G, Poli G, Francalanci M, Squecco R, Di Franco A, Borgogni E, Frontera S, Nesi G, Liotta F, Lucchese M, Perigli G, Francini F, Forti G, Serio M, Luconi M. Functional differences in visceral and subcutaneous fat pads originate from differences in the adipose stem cell. PLoS One 2012; 7:e36569. [PMID: 22574183 PMCID: PMC3344924 DOI: 10.1371/journal.pone.0036569] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 04/10/2012] [Indexed: 12/18/2022] Open
Abstract
Metabolic pathologies mainly originate from adipose tissue (AT) dysfunctions. AT differences are associated with fat-depot anatomic distribution in subcutaneous (SAT) and visceral omental (VAT) pads. We address the question whether the functional differences between the two compartments may be present early in the adipose stem cell (ASC) instead of being restricted to the mature adipocytes. Using a specific human ASC model, we evaluated proliferation/differentiation of ASC from abdominal SAT-(S-ASC) and VAT-(V-ASC) paired biopsies in parallel as well as the electrophysiological properties and functional activity of ASC and their in vitro-derived adipocytes. A dramatic difference in proliferation and adipogenic potential was observed between the two ASC populations, S-ASC having a growth rate and adipogenic potential significantly higher than V-ASC and giving rise to more functional and better organized adipocytes. To our knowledge, this is the first comprehensive electrophysiological analysis of ASC and derived-adipocytes, showing electrophysiological properties, such as membrane potential, capacitance and K(+)-current parameters which confirm the better functionality of S-ASC and their derived adipocytes. We document the greater ability of S-ASC-derived adipocytes to secrete adiponectin and their reduced susceptibility to lipolysis. These features may account for the metabolic differences observed between the SAT and VAT. Our findings suggest that VAT and SAT functional differences originate at the level of the adult ASC which maintains a memory of its fat pad of origin. Such stem cell differences may account for differential adipose depot susceptibility to the development of metabolic dysfunction and may represent a suitable target for specific therapeutic approaches.
Collapse
Affiliation(s)
- Silvana Baglioni
- Endocrine Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | - Giulia Cantini
- Endocrine Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | - Giada Poli
- Endocrine Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | - Michela Francalanci
- Endocrine Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | - Roberta Squecco
- Department of Physiological Sciences, University of Florence, Florence, Italy
| | - Alessandra Di Franco
- Endocrine Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | - Elisa Borgogni
- Endocrine Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | - Salvatore Frontera
- Endocrine Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | - Gabriella Nesi
- Department of Human Pathology and Oncology, University of Florence, Florence, Italy
| | - Francesco Liotta
- Department of Internal Medicine, University of Florence, Florence, Italy
| | | | - Giuliano Perigli
- Department of General Surgery, University of Florence, Florence, Italy
| | - Fabio Francini
- Department of Physiological Sciences, University of Florence, Florence, Italy
| | - Gianni Forti
- Endocrine Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | - Mario Serio
- Endocrine Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | - Michaela Luconi
- Endocrine Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
- * E-mail:
| |
Collapse
|
40
|
Karagiannides I, Bakirtzi K, Kokkotou E, Stavrakis D, Margolis KG, Thomou T, Giorgadze N, Kirkland JL, Pothoulakis C. Role of substance P in the regulation of glucose metabolism via insulin signaling-associated pathways. Endocrinology 2011; 152:4571-80. [PMID: 22009727 PMCID: PMC3230056 DOI: 10.1210/en.2011-1170] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Substance P (SP), encoded by the tachykinin 1 (Tac1) gene, is the most potent tachykinin ligand for the high-affinity neurokinin-1 receptor (NK-1R). We previously reported that NK-1R-deficient mice show less weight gain and reduced circulating levels of leptin and insulin in response to a high-fat diet (HFD) and demonstrated the presence of functional NK-1R in isolated human preadipocytes. Here we assessed the effects of SP on weight gain in response to HFD and determined glucose metabolism in Tac1-deficient (Tac1(-/-)) mice. The effect of SP on the expression of molecules that may predispose to reduced glucose uptake was also determined in isolated human mesenteric, omental, and sc preadipocytes. We show that although weight accumulation in response to HFD was similar between Tac1(-/-) mice and wild-type littermates, Tac1(-/-) mice demonstrated lower glucose and leptin and increased adiponectin blood levels and showed improved responses to insulin challenge after HFD. SP stimulated phosphorylation of c-Jun N-terminal kinase, protein kinase C, mammalian target of rapamycin, and inhibitory serine insulin receptor substrate-1 phosphorylation in human preadipocytes in vitro. Preincubation of human mesenteric preadipocytes with the protein kinase C pseudosubstrate inhibitor reduced insulin receptor substrate 1 phosphorylation in response to SP. Lastly, SP also induced insulin receptor substrate-1 phosphorylation in mature human sc adipocytes. Our results demonstrate an important role for SP in adipose tissue responses and obesity-associated pathologies. These novel SP effects on molecules that enhance insulin resistance at the adipocyte level may reflect an important role for this peptide in the pathophysiology of type 2 diabetes.
Collapse
Affiliation(s)
- Iordanes Karagiannides
- Inflammatory Bowel Disease Center, Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, 675 Charles E. Young Drive, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Filippin-Monteiro FB, de Oliveira EM, Sandri S, Knebel FH, Albuquerque RC, Campa A. Serum amyloid A is a growth factor for 3T3-L1 adipocytes, inhibits differentiation and promotes insulin resistance. Int J Obes (Lond) 2011; 36:1032-9. [PMID: 21986708 PMCID: PMC3419975 DOI: 10.1038/ijo.2011.193] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND/OBJECTIVES Serum amyloid A (SAA) is an acute-phase protein that has been recently correlated with obesity and insulin resistance. Therefore, we first examined whether human recombinant SAA (rSAA) could affect the proliferation, differentiation and metabolism of 3T3-L1 preadipocytes. DESIGN Preadipocytes were treated with rSAA and analyzed for changes in viability and [³H-methyl]-thymidine incorporation as well as cell cycle perturbations using flow cytometry analysis. The mRNA expression profiles of adipogenic factors during the differentiation protocol were also analyzed using real-time PCR. After differentiation, 2-deoxy-[1,2-³H]-glucose uptake and glycerol release were evaluated. RESULTS rSAA treatment caused a 2.6-fold increase in cell proliferation, which was consistent with the results from flow cytometry showing that rSAA treatment augmented the percentage of cells in the S phase (60.9±0.54%) compared with the control cells (39.8±2.2%, (***) P<0.001). The rSAA-induced cell proliferation was mediated by the ERK1/2 signaling pathway, which was assessed by pretreatment with the inhibitor PD98059. However, the exposure of 3T3-L1 cells to rSAA during the differentiation process resulted in attenuated adipogenesis and decreased expression of adipogenesis-related factors. During the first 72 h of differentiation, rSAA inhibited the differentiation process by altering the mRNA expression kinetics of adipogenic transcription factors and proteins, such as PPARγ2 (peroxisome proliferator-activated receptor γ 2), C/EBPβ (CCAAT/enhancer-binding protein β) and GLUT4. rSAA prevented the intracellular accumulation of lipids and, in fully differentiated cells, increased lipolysis and prevented 2-deoxy-[1,2-³H]-glucose uptake, which favors insulin resistance. Additionally, rSAA stimulated the secretion of proinflammatory cytokines interleukin 6 and tumor necrosis factor α, and upregulated SAA3 mRNA expression during adipogenesis. CONCLUSIONS We showed that rSAA enhanced proliferation and inhibited differentiation in 3T3-L1 preadipocytes and altered insulin sensitivity in differentiated cells. These results highlight the complex role of SAA in the adipogenic process and support a direct link between obesity and its co-morbidities such as type II diabetes.
Collapse
|
42
|
Roldan M, Macias-Gonzalez M, Garcia R, Tinahones FJ, Martin M. Obesity short-circuits stemness gene network in human adipose multipotent stem cells. FASEB J 2011; 25:4111-26. [PMID: 21846837 DOI: 10.1096/fj.10-171439] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The discovery of adipose multipotent stem cells has provided new insights to explore cellular mechanisms involved in adipose tissue function. In the present work, we aimed to evaluate how the adipogenic environment influences the stemness of the resident multipotent stem cells. To achieve this goal, human omental multipotent stem cells (hO-MSCs) were isolated, expanded, and characterized in both healthy lean and morbidly obese individuals. We observed decreased cell proliferation, premature senescence, and increased cytokine secretion associated with increasing body mass index of the patients. Consistent with the latter finding, the hO-MSCs derived from patients with morbid obesity lose their multilineage differentiation capacity associated with a dysregulation in the Wnt, Notch, and Sonic Hedgehog signaling pathways. Moreover, microRNAs involved in the regulation of stemness, cell differentiation, and senescence were also up-regulated in obese individuals. Altogether, our data show that obesity causes a general short circuit in the stemness gene network of hO-MSCs.
Collapse
Affiliation(s)
- Mar Roldan
- Fundacion Instituto Mediterráneo para el Avance de la Biotecnología y la Investigación Sanitaria (IMABIS), Malaga, Spain
| | | | | | | | | |
Collapse
|
43
|
Xiang X, Zhao J, Xu G, Li Y, Zhang W. mTOR and the differentiation of mesenchymal stem cells. Acta Biochim Biophys Sin (Shanghai) 2011; 43:501-10. [PMID: 21642276 DOI: 10.1093/abbs/gmr041] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The mammalian target of rapamycin (mTOR), an evolutionarily conserved serine-threonine protein kinase, belongs to the phosphoinositide 3-kinase (PI3K)-related kinase family, which contains a lipid kinase-like domain within their C-terminal region. Recent studies have revealed that mTOR as a critical intracellular molecule can sense the extracellular energy status and regulate the cell growth and proliferation in a variety of cells and tissues. This review summarizes our current understanding about the effects of mTOR on cell differentiation and tissue development, with an emphasis on the lineage determination of mesenchymal stem cells. mTOR can promote adipogenesis in white adipocytes, brown adipocytes, and muscle satellite cells, while rapamycin inhibits the adipogenic function of mTOR. mTOR signaling may function to affect osteoblast proliferation and differentiation, however, rapamycin has been reported to either inhibit or promote osteogenesis. Although the precise mechanism remains unclear, mTOR is indispensable for myogenesis. Depending on the cell type, rapamycin has been reported to inhibit, promote, or have no effect on myogenesis.
Collapse
Affiliation(s)
- Xinxin Xiang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Department of Physiology and Pathophysiology, Health Science Center, Peking University, Beijing, China
| | | | | | | | | |
Collapse
|