1
|
Zhang J, Ye C, Zhu Y, Wang J, Liu J. The Cell-Specific Role of SHP2 in Regulating Bone Homeostasis and Regeneration Niches. Int J Mol Sci 2023; 24:ijms24032202. [PMID: 36768520 PMCID: PMC9917188 DOI: 10.3390/ijms24032202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
Src homology-2 containing protein tyrosine phosphatase (SHP2), encoded by PTPN11, has been proven to participate in bone-related diseases, such as Noonan syndrome (NS), metachondromatosis and osteoarthritis. However, the mechanisms of SHP2 in bone remodeling and homeostasis maintenance are complex and undemonstrated. The abnormal expression of SHP2 can influence the differentiation and maturation of osteoblasts, osteoclasts and chondrocytes. Meanwhile, SHP2 mutations can act on the immune system, vasculature and nervous system, which in turn affect bone development and remodeling. Signaling pathways regulated by SHP2, such as mitogen-activated protein kinase (MAPK), Indian hedgehog (IHH) and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT), are also involved in the proliferation, differentiation and migration of bone functioning cells. This review summarizes the recent advances of SHP2 on osteogenesis-related cells and niche cells in the bone marrow microenvironment. The phenotypic features of SHP2 conditional knockout mice and underlying mechanisms are discussed. The prospective applications of the current agonists or inhibitors that target SHP2 in bone-related diseases are also described. Full clarification of the role of SHP2 in bone remodeling will shed new light on potential treatment for bone related diseases.
Collapse
Affiliation(s)
- Jie Zhang
- Laboratory for Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chengxinyue Ye
- Laboratory for Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yufan Zhu
- Laboratory for Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (J.W.); (J.L.)
| | - Jin Liu
- Laboratory for Aging Research, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Correspondence: (J.W.); (J.L.)
| |
Collapse
|
2
|
Population Structure and Selection Signatures Underlying Domestication Inferred from Genome-Wide Copy Number Variations in Chinese Indigenous Pigs. Genes (Basel) 2022; 13:genes13112026. [PMID: 36360263 PMCID: PMC9690591 DOI: 10.3390/genes13112026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 10/28/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Single nucleotide polymorphism was widely used to perform genetic and evolution research in pigs. However, little is known about the effect of copy number variation (CNV) on characteristics in pigs. This study performed a genome-wide comparison of CNVs between Wannan black pigs (WBP) and Asian wild boars (AWB), using whole genome resequencing data. By using Manta, we detected in total 28,720 CNVs that covered approximately 1.98% of the pig genome length. We identified 288 selected CNVs (top 1%) by performing Fst statistics. Functional enrichment analyses for genes located in selected CNVs were found to be muscle related (NDN, TMOD4, SFRP1, and SMYD3), reproduction related (GJA1, CYP26B1, WNT5A, SRD5A2, PTPN11, SPEF2, and CCNB1), residual feed intake (RFI) related (MAP3K5), and ear size related (WIF1). This study provides essential information on selected CNVs in Wannan black pigs for further research on the genetic basis of the complex phenotypic and provides essential information for direction in the protection and utilization of Wannan black pig.
Collapse
|
3
|
Bajia D, Bottani E, Derwich K. Effects of Noonan Syndrome-Germline Mutations on Mitochondria and Energy Metabolism. Cells 2022; 11:cells11193099. [PMID: 36231062 PMCID: PMC9563972 DOI: 10.3390/cells11193099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/21/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022] Open
Abstract
Noonan syndrome (NS) and related Noonan syndrome with multiple lentigines (NSML) contribute to the pathogenesis of human diseases in the RASopathy family. This family of genetic disorders constitute one of the largest groups of developmental disorders with variable penetrance and severity, associated with distinctive congenital disabilities, including facial features, cardiopathies, growth and skeletal abnormalities, developmental delay/mental retardation, and tumor predisposition. NS was first clinically described decades ago, and several genes have since been identified, providing a molecular foundation to understand their physiopathology and identify targets for therapeutic strategies. These genes encode proteins that participate in, or regulate, RAS/MAPK signalling. The RAS pathway regulates cellular metabolism by controlling mitochondrial homeostasis, dynamics, and energy production; however, little is known about the role of mitochondrial metabolism in NS and NSML. This manuscript comprehensively reviews the most frequently mutated genes responsible for NS and NSML, covering their role in the current knowledge of cellular signalling pathways, and focuses on the pathophysiological outcomes on mitochondria and energy metabolism.
Collapse
Affiliation(s)
- Donald Bajia
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Ul. Fredry 10, 61701 Poznan, Poland
| | - Emanuela Bottani
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona, Piazzale L. A. Scuro 10, 37134 Verona, Italy
- Correspondence: (E.B.); (K.D.); Tel.: +39-3337149584 (E.B.); +48-504199285 (K.D.)
| | - Katarzyna Derwich
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Ul. Fredry 10, 61701 Poznan, Poland
- Correspondence: (E.B.); (K.D.); Tel.: +39-3337149584 (E.B.); +48-504199285 (K.D.)
| |
Collapse
|
4
|
The Tyrosine Phosphatase SHP2: A New Target for Insulin Resistance? Biomedicines 2022; 10:biomedicines10092139. [PMID: 36140242 PMCID: PMC9495760 DOI: 10.3390/biomedicines10092139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/26/2022] [Accepted: 08/28/2022] [Indexed: 11/17/2022] Open
Abstract
The SH2 containing protein tyrosine phosphatase 2(SHP2) plays essential roles in fundamental signaling pathways, conferring on it versatile physiological functions during development and in homeostasis maintenance, and leading to major pathological outcomes when dysregulated. Many studies have documented that SHP2 modulation disrupted glucose homeostasis, pointing out a relationship between its dysfunction and insulin resistance, and the therapeutic potential of its targeting. While studies from cellular or tissue-specific models concluded on both pros-and-cons effects of SHP2 on insulin resistance, recent data from integrated systems argued for an insulin resistance promoting role for SHP2, and therefore a therapeutic benefit of its inhibition. In this review, we will summarize the general knowledge of SHP2’s molecular, cellular, and physiological functions, explaining the pathophysiological impact of its dysfunctions, then discuss its protective or promoting roles in insulin resistance as well as the potency and limitations of its pharmacological modulation.
Collapse
|
5
|
Zhao Z, Bai Y, Tian H, Shi B, Li X, Luo Y, Wang J, Hu J, Abbas Raza SH. Interference with ACSL1 gene in bovine adipocytes: Transcriptome profiling of circRNA related to unsaturated fatty acid production. Genomics 2021; 113:3967-3977. [PMID: 34601049 DOI: 10.1016/j.ygeno.2021.09.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 07/23/2021] [Accepted: 09/27/2021] [Indexed: 01/12/2023]
Abstract
Long-chain acyl-CoA synthetase 1 (ACSL1) is a member of the acyl-CoA synthetase family that plays a vital role in lipid metabolism. We have previously shown that the ACSL1 gene regulates the composition of unsaturated fatty acids (UFAs) in bovine skeletal muscle, which in turn regulates the fatty acid synthesis and the generation of lipid droplets. Here, we used RNA-Seq to screen circRNAs that regulated the expression of ACSL1 gene and other UFA synthesis-related genes by RNA interference and noninterference in bovine adipocytes. The results of KEGG pathway analysis showed that the parental genes of differentially expressed (DE)-circRNAs were primarily enriched in the adipocytokine signaling pathway. The prediction results showed that novel_circ_0004855, novel_circ_0001507, novel_circ_0001731, novel_circ_0005276, novel_circ_0002060, novel_circ_0005405 and novel_circ_0004254 regulated UFA synthesis-related genes by interacting with the related miRNAs. These results could help expand our knowledge of the molecular mechanisms of circRNAs in the regulation of UFA synthesis in bovine adipocytes.
Collapse
Affiliation(s)
- Zhidong Zhao
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Yanbin Bai
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Hongshan Tian
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Bingang Shi
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Xupeng Li
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Yuzhu Luo
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jiqing Wang
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jiang Hu
- College of Animal Science and Technology, Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China.
| | - Sayed Haidar Abbas Raza
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
6
|
Paccoud R, Saint-Laurent C, Piccolo E, Tajan M, Dortignac A, Pereira O, Le Gonidec S, Baba I, Gélineau A, Askia H, Branchereau M, Charpentier J, Personnaz J, Branka S, Auriau J, Deleruyelle S, Canouil M, Beton N, Salles JP, Tauber M, Weill J, Froguel P, Neel BG, Araki T, Heymes C, Burcelin R, Castan I, Valet P, Dray C, Gautier EL, Edouard T, Pradère JP, Yart A. SHP2 drives inflammation-triggered insulin resistance by reshaping tissue macrophage populations. Sci Transl Med 2021; 13:13/591/eabe2587. [PMID: 33910978 DOI: 10.1126/scitranslmed.abe2587] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 04/05/2021] [Indexed: 12/11/2022]
Abstract
Insulin resistance is a key event in type 2 diabetes onset and a major comorbidity of obesity. It results from a combination of fat excess-triggered defects, including lipotoxicity and metaflammation, but the causal mechanisms remain difficult to identify. Here, we report that hyperactivation of the tyrosine phosphatase SHP2 found in Noonan syndrome (NS) led to an unsuspected insulin resistance profile uncoupled from altered lipid management (for example, obesity or ectopic lipid deposits) in both patients and mice. Functional exploration of an NS mouse model revealed this insulin resistance phenotype correlated with constitutive inflammation of tissues involved in the regulation of glucose metabolism. Bone marrow transplantation and macrophage depletion improved glucose homeostasis and decreased metaflammation in the mice, highlighting a key role of macrophages. In-depth analysis of bone marrow-derived macrophages in vitro and liver macrophages showed that hyperactive SHP2 promoted a proinflammatory phenotype, modified resident macrophage homeostasis, and triggered monocyte infiltration. Consistent with a role of SHP2 in promoting inflammation-driven insulin resistance, pharmaceutical SHP2 inhibition in obese diabetic mice improved insulin sensitivity even better than conventional antidiabetic molecules by specifically reducing metaflammation and alleviating macrophage activation. Together, these results reveal that SHP2 hyperactivation leads to inflammation-triggered metabolic impairments and highlight the therapeutical potential of SHP2 inhibition to ameliorate insulin resistance.
Collapse
Affiliation(s)
- Romain Paccoud
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France
| | - Céline Saint-Laurent
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France.,RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse F-31100, France
| | - Enzo Piccolo
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France.,RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse F-31100, France
| | - Mylène Tajan
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France.,RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse F-31100, France
| | - Alizée Dortignac
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France
| | - Ophélie Pereira
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France.,RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse F-31100, France
| | - Sophie Le Gonidec
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France.,RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse F-31100, France
| | - Inès Baba
- INSERM UMR-S 1166, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris F-75013, France
| | - Adélaïde Gélineau
- INSERM UMR-S 1166, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris F-75013, France
| | - Haoussa Askia
- INSERM UMR-S 1166, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris F-75013, France
| | - Maxime Branchereau
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France
| | - Julie Charpentier
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France
| | - Jean Personnaz
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France
| | - Sophie Branka
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France
| | - Johanna Auriau
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France
| | - Simon Deleruyelle
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France
| | - Mickaël Canouil
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille University Hospital, Lille F-59000, France
| | - Nicolas Beton
- Endocrine, Bone Diseases, and Genetics Unit, Children's Hospital, Toulouse University Hospital, Toulouse France and Centre de Physiopathologie Toulouse-Purpan, INSERM UMR 1043, Université Paul Sabatier, Université de Toulouse, Toulouse F-31024, France
| | - Jean-Pierre Salles
- Endocrine, Bone Diseases, and Genetics Unit, Children's Hospital, Toulouse University Hospital, Toulouse France and Centre de Physiopathologie Toulouse-Purpan, INSERM UMR 1043, Université Paul Sabatier, Université de Toulouse, Toulouse F-31024, France
| | - Maithé Tauber
- Endocrine, Bone Diseases, and Genetics Unit, Children's Hospital, Toulouse University Hospital, Toulouse France and Centre de Physiopathologie Toulouse-Purpan, INSERM UMR 1043, Université Paul Sabatier, Université de Toulouse, Toulouse F-31024, France
| | - Jacques Weill
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille University Hospital, Lille F-59000, France
| | - Philippe Froguel
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, University of Lille, Lille University Hospital, Lille F-59000, France.,Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
| | - Benjamin G Neel
- Laura and Isaac Perlmutter Cancer Center, NYU-Langone Medical Center, NY 10016, USA
| | - Toshiyuki Araki
- Laura and Isaac Perlmutter Cancer Center, NYU-Langone Medical Center, NY 10016, USA
| | - Christophe Heymes
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France
| | - Rémy Burcelin
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France
| | - Isabelle Castan
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France.,RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse F-31100, France
| | - Philippe Valet
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France.,RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse F-31100, France
| | - Cédric Dray
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France.,RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse F-31100, France
| | - Emmanuel L Gautier
- INSERM UMR-S 1166, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris F-75013, France
| | - Thomas Edouard
- RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse F-31100, France.,Endocrine, Bone Diseases, and Genetics Unit, Children's Hospital, Toulouse University Hospital, Toulouse France and Centre de Physiopathologie Toulouse-Purpan, INSERM UMR 1043, Université Paul Sabatier, Université de Toulouse, Toulouse F-31024, France
| | - Jean-Philippe Pradère
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France.,RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse F-31100, France
| | - Armelle Yart
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, Université Paul Sabatier, Université de Toulouse, Toulouse F-31432, France. .,RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse F-31100, France
| |
Collapse
|
7
|
Yang L, Han X, Zhang C, Sun C, Huang S, Xiao W, Gao Y, Liang Q, Luo F, Lu W, Fu J, Zhou Y. Hsa_circ_0060450 Negatively Regulates Type I Interferon-Induced Inflammation by Serving as miR-199a-5p Sponge in Type 1 Diabetes Mellitus. Front Immunol 2020; 11:576903. [PMID: 33133095 PMCID: PMC7550460 DOI: 10.3389/fimmu.2020.576903] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 08/31/2020] [Indexed: 12/15/2022] Open
Abstract
Circular RNAs (circRNAs) constitute a class of covalently circular non-coding RNA molecules formed by 5′ and 3′ end back-splicing. The rapid development of bioinformatics and large-scale sequencing has led to the identification of functional circRNAs. Despite an overall upward trend, studies focusing on the roles of circRNAs in immune diseases remain relatively scarce. In the present study, we obtained a differential circRNA expression profile based on microarray analysis of peripheral blood mononuclear cells (PBMCs) in children with type 1 diabetes mellitus (T1DM). We characterized one differentially expressed circRNA back-spliced from the MYB Proto-Oncogene Like 2 (MYBL2) gene in patients with T1DM, termed as hsa_circ_0060450. Subsequent assays revealed that hsa_circ_0060450 can serve as the sponge of miR-199a-5p, release its target gene, Src homology 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2), encoded by the tyrosine-protein phosphatase non-receptor type 11 gene (PTPN11), and further suppress the JAK-STAT signaling pathway triggered by type I interferon (IFN-I) to inhibit macrophage-mediated inflammation, which indicates the important roles of circRNAs in T1DM and represents a promising therapeutic molecule in the treatment of T1DM.
Collapse
Affiliation(s)
- Lan Yang
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| | - Xiao Han
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| | - Caiyan Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| | - Chengjun Sun
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Saihua Huang
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| | - Wenfeng Xiao
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| | - Yajing Gao
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| | - Qiuyan Liang
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| | - Feihong Luo
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Wei Lu
- Department of Pediatric Endocrinology and Inherited Metabolic Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Jinrong Fu
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yufeng Zhou
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,National Health Commission (NHC) Key Laboratory of Neonatal Diseases, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Liu M, Jin HS, Park S. Protein and fat intake interacts with the haplotype of PTPN11_rs11066325, RPH3A_rs886477, and OAS3_rs2072134 to modulate serum HDL concentrations in middle-aged people. Clin Nutr 2020; 39:942-949. [PMID: 31006500 DOI: 10.1016/j.clnu.2019.03.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Low serum HDL cholesterol (HDL-C) concentration is a risk factor for cardiovascular diseases and it is influenced by genetic and environmental factors. We hypothesized that genetic variants that decrease serum HDL-C concentrations may interact with nutrient intakes in ways that increase or decrease the risk of cardiovascular disease. METHODS Candidate genetic variants that can lower serum HDL-C concentrations were explored by genome-wide association studies (GWAS), after adjusting for covariates, in the Ansan/Ansung cohort (n = 8842) from KoGES. The best genetic variants were selected and used to form a haplotype. According to the haplotype frequencies of SNPs, they were divided into major allele, heterozygote allele, and minor allele. The association of haplotype with serum HDL-C levels was determined using logistic regression after adjusting for confounding factors. Interaction of the haplotype with nutrient intake was also determined. RESULTS PTPN11_rs11066325, RPH3A_rs886477 and OAS3_rs2072134 were selected to modulate serum HDL-C levels from GWAS(P = 1.09E-09, 7.04E-10, and 1.27E-09, respectively). The adjusted odds ratios (ORs) for a decrease in serum HDL-C concentration in the minor-allele group of the haplotype were elevated by 1.534 fold, compared to the major-allele group of the haplotype. Furthermore, the adjusted ORs for serum LDL cholesterol and levels increased by 1.645 in the minor-alleles compared to the major-alleles of the haplotype without a significant change of serum cholesterol levels. Interestingly, the adjusted ORs for serum triglyceride were lower in the minor-alleles than in the major-alleles. The haplotype had a significant interaction with the intake of protein, fat, saturated fatty acids (SAF) and polyunsaturated fatty acids (PUFA; P < 0.05). In particular, the minor alleles of the haplotype decreased serum HDL-C levels compared to the major-alleles in the high intake of protein, fat, SFA, and PUFA, not in the low intake. CONCLUSIONS People carrying the minor-allele of haplotypes should avoid diets that are high in protein and fat, especially rich in SFA and PUFA.
Collapse
Affiliation(s)
- Meiling Liu
- Dept. of Food and Nutrition, Institue of Basic Science, Obesity/Diabetes Research Center, Hoseo University, Asan, Chungnam, 31499, South Korea
| | - Hyun Seok Jin
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, Asan, Chungnam, 31499, South Korea
| | - Sunmin Park
- Dept. of Food and Nutrition, Institue of Basic Science, Obesity/Diabetes Research Center, Hoseo University, Asan, Chungnam, 31499, South Korea.
| |
Collapse
|
9
|
Hall JE, do Carmo JM, da Silva AA, Wang Z, Hall ME. Obesity, kidney dysfunction and hypertension: mechanistic links. Nat Rev Nephrol 2020; 15:367-385. [PMID: 31015582 DOI: 10.1038/s41581-019-0145-4] [Citation(s) in RCA: 304] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Excessive adiposity raises blood pressure and accounts for 65-75% of primary hypertension, which is a major driver of cardiovascular and kidney diseases. In obesity, abnormal kidney function and associated increases in tubular sodium reabsorption initiate hypertension, which is often mild before the development of target organ injury. Factors that contribute to increased sodium reabsorption in obesity include kidney compression by visceral, perirenal and renal sinus fat; increased renal sympathetic nerve activity (RSNA); increased levels of anti-natriuretic hormones, such as angiotensin II and aldosterone; and adipokines, particularly leptin. The renal and neurohormonal pathways of obesity and hypertension are intertwined. For example, leptin increases RSNA by stimulating the central nervous system proopiomelanocortin-melanocortin 4 receptor pathway, and kidney compression and RSNA contribute to renin-angiotensin-aldosterone system activation. Glucocorticoids and/or oxidative stress may also contribute to mineralocorticoid receptor activation in obesity. Prolonged obesity and progressive renal injury often lead to the development of treatment-resistant hypertension. Patient management therefore often requires multiple antihypertensive drugs and concurrent treatment of dyslipidaemia, insulin resistance, diabetes and inflammation. If more effective strategies for the prevention and control of obesity are not developed, cardiorenal, metabolic and other obesity-associated diseases could overwhelm health-care systems in the future.
Collapse
Affiliation(s)
- John E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA. .,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Jussara M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alexandre A da Silva
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Zhen Wang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
10
|
Cohen-Sharir Y, Kuperman Y, Apelblat D, den Hertog J, Spiegel I, Knobler H, Elson A. Protein tyrosine phosphatase alpha inhibits hypothalamic leptin receptor signaling and regulates body weight in vivo. FASEB J 2019; 33:5101-5111. [PMID: 30615487 DOI: 10.1096/fj.201800860rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Understanding how body weight is regulated at the molecular level is essential for treating obesity. We show that female mice genetically lacking protein tyrosine phosphatase (PTP) receptor type α (PTPRA) exhibit reduced weight and adiposity and increased energy expenditure, and are more resistant to diet-induced obesity than matched wild-type control mice. These mice also exhibit reduced levels of circulating leptin and are leptin hypersensitive, suggesting that PTPRA inhibits leptin signaling in the hypothalamus. Male and female PTPRA-deficient mice fed a high-fat diet were leaner and displayed increased metabolic rates and lower circulating leptin levels, indicating that the effects of loss of PTPRA persist in the obese state. Molecularly, PTPRA down-regulates leptin receptor signaling by dephosphorylating the receptor-associated kinase JAK2, with which the phosphatase associates constitutively. In contrast to the closely related tyrosine phosphatase ε, leptin induces only weak phosphorylation of PTPRA at its C-terminal regulatory site Y789, and this does not affect the activity of PTPRA toward JAK2. PTPRA is therefore an inhibitor of hypothalamic leptin signaling in vivo and may prevent premature activation of leptin signaling, as well as return signaling to baseline after exposure to leptin.-Cohen-Sharir, Y., Kuperman, Y., Apelblat, D., den Hertog, J., Spiegel, I., Knobler, H., Elson, A. Protein tyrosine phosphatase alpha inhibits hypothalamic leptin receptor signaling and regulates body weight in vivo.
Collapse
Affiliation(s)
- Yael Cohen-Sharir
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Daniella Apelblat
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Jeroen den Hertog
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht, The Netherlands.,Institute Biology Leiden, Leiden, The Netherlands; and
| | - Ivo Spiegel
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Hilla Knobler
- Diabetes, Endocrinology and Metabolic Institute, Kaplan Medical Center, Rehovot, Israel
| | - Ari Elson
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
11
|
Leptin Signaling in the Control of Metabolism and Appetite: Lessons from Animal Models. J Mol Neurosci 2018; 66:390-402. [DOI: 10.1007/s12031-018-1185-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/24/2018] [Indexed: 12/15/2022]
|
12
|
Moreira GCM, Boschiero C, Cesar ASM, Reecy JM, Godoy TF, Trevisoli PA, Cantão ME, Ledur MC, Ibelli AMG, Peixoto JDO, Moura ASAMT, Garrick D, Coutinho LL. A genome-wide association study reveals novel genomic regions and positional candidate genes for fat deposition in broiler chickens. BMC Genomics 2018; 19:374. [PMID: 29783939 PMCID: PMC5963092 DOI: 10.1186/s12864-018-4779-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/10/2018] [Indexed: 12/21/2022] Open
Abstract
Background Excess fat content in chickens has a negative impact on poultry production. The discovery of QTL associated with fat deposition in the carcass allows the identification of positional candidate genes (PCGs) that might regulate fat deposition and be useful for selection against excess fat content in chicken’s carcass. This study aimed to estimate genomic heritability coefficients and to identify QTLs and PCGs for abdominal fat (ABF) and skin (SKIN) traits in a broiler chicken population, originated from the White Plymouth Rock and White Cornish breeds. Results ABF and SKIN are moderately heritable traits in our broiler population with estimates ranging from 0.23 to 0.33. Using a high density SNP panel (355,027 informative SNPs), we detected nine unique QTLs that were associated with these fat traits. Among these, four QTL were novel, while five have been previously reported in the literature. Thirteen PCGs were identified that might regulate fat deposition in these QTL regions: JDP2, PLCG1, HNF4A, FITM2, ADIPOR1, PTPN11, MVK, APOA1, APOA4, APOA5, ENSGALG00000000477, ENSGALG00000000483, and ENSGALG00000005043. We used sequence information from founder animals to detect 4843 SNPs in the 13 PCGs. Among those, two were classified as potentially deleterious and two as high impact SNPs. Conclusions This study generated novel results that can contribute to a better understanding of fat deposition in chickens. The use of high density array of SNPs increases genome coverage and improves QTL resolution than would have been achieved with low density. The identified PCGs were involved in many biological processes that regulate lipid storage. The SNPs identified in the PCGs, especially those predicted as potentially deleterious and high impact, may affect fat deposition. Validation should be undertaken before using these SNPs for selection against carcass fat accumulation and to improve feed efficiency in broiler chicken production. Electronic supplementary material The online version of this article (10.1186/s12864-018-4779-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gabriel Costa Monteiro Moreira
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Piracicaba, São Paulo, 13418-900, Brazil
| | - Clarissa Boschiero
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Piracicaba, São Paulo, 13418-900, Brazil
| | - Aline Silva Mello Cesar
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Piracicaba, São Paulo, 13418-900, Brazil
| | - James M Reecy
- Department of Animal Science, Iowa State University (ISU), Ames, Iowa, USA
| | - Thaís Fernanda Godoy
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Piracicaba, São Paulo, 13418-900, Brazil
| | - Priscila Anchieta Trevisoli
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Piracicaba, São Paulo, 13418-900, Brazil
| | | | | | | | | | | | - Dorian Garrick
- School of Agriculture, Massey University, Ruakura, Hamilton, New Zealand
| | - Luiz Lehmann Coutinho
- Department of Animal Science, University of São Paulo (USP) / Luiz de Queiroz College of Agriculture (ESALQ), Piracicaba, São Paulo, 13418-900, Brazil.
| |
Collapse
|
13
|
Abstract
The RAS/MAPK signaling pathway plays key roles in development, cell survival and proliferation, as well as in cancer pathogenesis. Molecular genetic studies have identified a group of developmental syndromes, the RASopathies, caused by germ line mutations in this pathway. The syndromes included within this classification are neurofibromatosis type 1 (NF1), Noonan syndrome (NS), Noonan syndrome with multiple lentigines (NS-ML, formerly known as LEOPARD syndrome), Costello syndrome (CS), cardio-facio-cutaneous syndrome (CFC), Legius syndrome (LS, NF1-like syndrome), capillary malformation-arteriovenous malformation syndrome (CM-AVM), and hereditary gingival fibromatosis (HGF) type 1. Although these syndromes present specific molecular alterations, they are characterized by a large spectrum of functional and morphological abnormalities, which include heart defects, short stature, neurocognitive impairment, craniofacial malformations, and, in some cases, cancer predisposition. The development of genetically modified animals, such as mice (Mus musculus), flies (Drosophila melanogaster), and zebrafish (Danio rerio), has been instrumental in elucidating the molecular and cellular bases of these syndromes. Moreover, these models can also be used to determine tumor predisposition, the impact of different genetic backgrounds on the variable phenotypes found among the patients and to evaluate preventative and therapeutic strategies. Here, we review a wide range of genetically modified mouse models used in the study of RASopathies and the potential application of novel technologies, which hopefully will help us resolve open questions in the field.
Collapse
|
14
|
Shpakov AO. The brain leptin signaling system and its functional state in metabolic syndrome and type 2 diabetes mellitus. J EVOL BIOCHEM PHYS+ 2016. [DOI: 10.1134/s0022093016030017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
15
|
SHP2 sails from physiology to pathology. Eur J Med Genet 2015; 58:509-25. [PMID: 26341048 DOI: 10.1016/j.ejmg.2015.08.005] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/24/2015] [Accepted: 08/30/2015] [Indexed: 02/08/2023]
Abstract
Over the two past decades, mutations of the PTPN11 gene, encoding the ubiquitous protein tyrosine phosphatase SHP2 (SH2 domain-containing tyrosine phosphatase 2), have been identified as the causal factor of several developmental diseases (Noonan syndrome (NS), Noonan syndrome with multiple lentigines (NS-ML), and metachondromatosis), and malignancies (juvenile myelomonocytic leukemia). SHP2 plays essential physiological functions in organism development and homeostasis maintenance by regulating fundamental intracellular signaling pathways in response to a wide range of growth factors and hormones, notably the pleiotropic Ras/Mitogen-Activated Protein Kinase (MAPK) and the Phosphoinositide-3 Kinase (PI3K)/AKT cascades. Analysis of the biochemical impacts of PTPN11 mutations first identified both loss-of-function and gain-of-function mutations, as well as more subtle defects, highlighting the major pathophysiological consequences of SHP2 dysregulation. Then, functional genetic studies provided insights into the molecular dysregulations that link SHP2 mutants to the development of specific traits of the diseases, paving the way for the design of specific therapies for affected patients. In this review, we first provide an overview of SHP2's structure and regulation, then describe its molecular roles, notably its functions in modulating the Ras/MAPK and PI3K/AKT signaling pathways, and its physiological roles in organism development and homeostasis. In the second part, we describe the different PTPN11 mutation-associated pathologies and their clinical manifestations, with particular focus on the biochemical and signaling outcomes of NS and NS-ML-associated mutations, and on the recent advances regarding the pathophysiology of these diseases.
Collapse
|
16
|
Hall JE, do Carmo JM, da Silva AA, Wang Z, Hall ME. Obesity-induced hypertension: interaction of neurohumoral and renal mechanisms. Circ Res 2015; 116:991-1006. [PMID: 25767285 DOI: 10.1161/circresaha.116.305697] [Citation(s) in RCA: 734] [Impact Index Per Article: 81.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Excess weight gain, especially when associated with increased visceral adiposity, is a major cause of hypertension, accounting for 65% to 75% of the risk for human primary (essential) hypertension. Increased renal tubular sodium reabsorption impairs pressure natriuresis and plays an important role in initiating obesity hypertension. The mediators of abnormal kidney function and increased blood pressure during development of obesity hypertension include (1) physical compression of the kidneys by fat in and around the kidneys, (2) activation of the renin-angiotensin-aldosterone system, and (3) increased sympathetic nervous system activity. Activation of the renin-angiotensin-aldosterone system is likely due, in part, to renal compression, as well as sympathetic nervous system activation. However, obesity also causes mineralocorticoid receptor activation independent of aldosterone or angiotensin II. The mechanisms for sympathetic nervous system activation in obesity have not been fully elucidated but may require leptin and activation of the brain melanocortin system. With prolonged obesity and development of target organ injury, especially renal injury, obesity-associated hypertension becomes more difficult to control, often requiring multiple antihypertensive drugs and treatment of other risk factors, including dyslipidemia, insulin resistance and diabetes mellitus, and inflammation. Unless effective antiobesity drugs are developed, the effect of obesity on hypertension and related cardiovascular, renal and metabolic disorders is likely to become even more important in the future as the prevalence of obesity continues to increase.
Collapse
Affiliation(s)
- John E Hall
- From the Departments of Physiology and Biophysics (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), Medicine (M.E.H.), Mississippi Center for Obesity Research (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), and Cardiovascular-Renal Research Center (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), University of Mississippi Medical Center, Jackson.
| | - Jussara M do Carmo
- From the Departments of Physiology and Biophysics (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), Medicine (M.E.H.), Mississippi Center for Obesity Research (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), and Cardiovascular-Renal Research Center (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), University of Mississippi Medical Center, Jackson
| | - Alexandre A da Silva
- From the Departments of Physiology and Biophysics (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), Medicine (M.E.H.), Mississippi Center for Obesity Research (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), and Cardiovascular-Renal Research Center (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), University of Mississippi Medical Center, Jackson
| | - Zhen Wang
- From the Departments of Physiology and Biophysics (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), Medicine (M.E.H.), Mississippi Center for Obesity Research (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), and Cardiovascular-Renal Research Center (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), University of Mississippi Medical Center, Jackson
| | - Michael E Hall
- From the Departments of Physiology and Biophysics (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), Medicine (M.E.H.), Mississippi Center for Obesity Research (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), and Cardiovascular-Renal Research Center (J.E.H., J.M.d.C., A.A.d.S., Z.W., M.E.H.), University of Mississippi Medical Center, Jackson
| |
Collapse
|
17
|
Bassi M, Furuya WI, Zoccal DB, Menani JV, Colombari E, Hall JE, da Silva AA, do Carmo JM, Colombari DSA. Control of respiratory and cardiovascular functions by leptin. Life Sci 2015; 125:25-31. [PMID: 25645056 PMCID: PMC4355938 DOI: 10.1016/j.lfs.2015.01.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/19/2014] [Accepted: 01/23/2015] [Indexed: 01/29/2023]
Abstract
Leptin, a peptide hormone produced by adipose tissue, acts in brain centers that control critical physiological functions such as metabolism, breathing and cardiovascular regulation. The importance of leptin for respiratory control is evident by the fact that leptin deficient mice exhibit impaired ventilatory responses to carbon dioxide (CO2), which can be corrected by intracerebroventricular leptin replacement therapy. Leptin is also recognized as an important link between obesity and hypertension. Humans and animal models lacking either leptin or functional leptin receptors exhibit many characteristics of the metabolic syndrome, including hyperinsulinemia, insulin resistance, hyperglycemia, dyslipidemia and visceral adiposity, but do not exhibit increased sympathetic nerve activity (SNA) and have normal to lower blood pressure (BP) compared to lean controls. Even though previous studies have extensively focused on the brain sites and intracellular signaling pathways involved in leptin effects on food intake and energy balance, the mechanisms that mediate the actions of leptin on breathing and cardiovascular function are only beginning to be elucidated. This mini-review summarizes recent advances on the effects of leptin on cardiovascular and respiratory control with emphasis on the neural control of respiratory function and autonomic activity.
Collapse
Affiliation(s)
- M Bassi
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil.
| | - W I Furuya
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - D B Zoccal
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - J V Menani
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - E Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - J E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - A A da Silva
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - J M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - D S A Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, SP, Brazil
| |
Collapse
|
18
|
Shp2 in forebrain neurons regulates synaptic plasticity, locomotion, and memory formation in mice. Mol Cell Biol 2015; 35:1557-72. [PMID: 25713104 DOI: 10.1128/mcb.01339-14] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/17/2015] [Indexed: 11/20/2022] Open
Abstract
Shp2 (Src homology 2 domain-containing protein tyrosine phosphatase 2) regulates neural cell differentiation. It is also expressed in postmitotic neurons, however, and mutations of Shp2 are associated with clinical syndromes characterized by mental retardation. Here we show that conditional-knockout (cKO) mice lacking Shp2 specifically in postmitotic forebrain neurons manifest abnormal behavior, including hyperactivity. Novelty-induced expression of immediate-early genes and activation of extracellular-signal-regulated kinase (Erk) were attenuated in the cerebral cortex and hippocampus of Shp2 cKO mice, suggestive of reduced neuronal activity. In contrast, ablation of Shp2 enhanced high-K(+)-induced Erk activation in both cultured cortical neurons and synaptosomes, whereas it inhibited that induced by brain-derived growth factor in cultured neurons. Posttetanic potentiation and paired-pulse facilitation were attenuated and enhanced, respectively, in hippocampal slices from Shp2 cKO mice. The mutant mice also manifested transient impairment of memory formation in the Morris water maze. Our data suggest that Shp2 contributes to regulation of Erk activation and synaptic plasticity in postmitotic forebrain neurons and thereby controls locomotor activity and memory formation.
Collapse
|
19
|
Gurzov EN, Stanley WJ, Brodnicki TC, Thomas HE. Protein tyrosine phosphatases: molecular switches in metabolism and diabetes. Trends Endocrinol Metab 2015; 26:30-9. [PMID: 25432462 DOI: 10.1016/j.tem.2014.10.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 02/06/2023]
Abstract
Protein tyrosine phosphatases (PTPs) are a large family of enzymes that generally oppose the actions of protein tyrosine kinases (PTKs). Genetic polymorphisms for particular PTPs are associated with altered risk of both type 1 diabetes (T1D) and type 2 diabetes (T2D). Moreover, recent evidence suggests that PTPs play crucial roles in metabolism. They can act as regulators of liver homeostasis, food intake, or immune-mediated pancreatic b cell death. In this review we describe the mechanisms by which different members of the non-receptor PTP (PTPN) family influence metabolic physiology. This 'metabolic job' of PTPs is discussed in depth and the role of these proteins in different cell types compared. Understanding the pathways regulated by PTPs will provide novel therapeutic strategies for the treatment of diabetes.
Collapse
|
20
|
Borges BDC, Rorato RC, Uchoa ET, Marangon PB, Elias CF, Antunes-Rodrigues J, Elias LLK. Protein tyrosine phosphatase-1B contributes to LPS-induced leptin resistance in male rats. Am J Physiol Endocrinol Metab 2015; 308:E40-50. [PMID: 25352433 PMCID: PMC4280212 DOI: 10.1152/ajpendo.00094.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 10/24/2014] [Indexed: 12/18/2022]
Abstract
Leptin resistance is induced by the feedback inhibitors tyrosine phosphatase-1B (PTP1B) and decreased Src homology 2 domain-containing tyrosine phosphatase-2 (SHP-2) signaling. To investigate the participation of PTP1B and SHP-2 in LPS-induced leptin resistance, we injected repeated (6-LPS) intraperitoneal LPS doses (100 μg/kg ip) for comparison with a single (1-LPS) treatment and evaluated the expression of SHP-2, PTP1B, p-ERK1/2, and p-STAT3 in the hypothalamus of male Wistar rats. The single LPS treatment increased the expression of p-STAT3 and PTP1B but not SHP-2. The repeated LPS treatment reduced SHP-2, increased PTP1B, and did not change p-STAT3. We observed that the PTP1B expression induced by the endotoxin was highly colocalized with leptin receptor cells in the hypothalamus of LepRb-IRES-Cre-tdTomato reporter mice. The single, but not the repeated, LPS treatment decreased the food intake and body weight. Leptin had no stimulatory effect on the hypophagia, body weight loss, or pSTAT3 expression in 6-LPS rats, indicating leptin unresponsiveness. Notably, the PTP1B inhibitor (3.0 nmol/rat in 5 μl icv) restored the LPS-induced hypophagia in 6-LPS rats and restored the ability of leptin to reduce food intake and body weight as well as to phosphorylate STAT3 in the arcuate, paraventricular, and ventromedial nuclei of the hypothalamus. The present data suggest that an increased PTP1B expression in the hypothalamus underlies the development of leptin resistance during repeated exposure to LPS. Our findings contribute to understanding the mechanisms involved in leptin resistance during low-grade inflammation as seen in obesity.
Collapse
Affiliation(s)
| | - Rodrigo C Rorato
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil; and
| | - Ernane Torres Uchoa
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil; and
| | - Paula B Marangon
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil; and
| | - Carol F Elias
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Jose Antunes-Rodrigues
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil; and
| | - Lucila L K Elias
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil; and
| |
Collapse
|
21
|
do Carmo JM, da Silva AA, Ebaady SE, Sessums PO, Abraham RS, Elmquist JK, Lowell BB, Hall JE. Shp2 signaling in POMC neurons is important for leptin's actions on blood pressure, energy balance, and glucose regulation. Am J Physiol Regul Integr Comp Physiol 2014; 307:R1438-47. [PMID: 25339680 DOI: 10.1152/ajpregu.00131.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Previous studies showed that Src homology-2 tyrosine phosphatase (Shp2) is an important regulator of body weight. In this study, we examined the impact of Shp2 deficiency specifically in proopiomelanocortin (POMC) neurons on metabolic and cardiovascular function and on chronic blood pressure (BP) and metabolic responses to leptin. Mice with Shp2 deleted in POMC neurons (Shp2/Pomc-cre) and control mice (Shp2(flox/flox)) were implanted with telemetry probes and venous catheters for measurement of mean arterial pressure (MAP) and leptin infusion. After at least 5 days of stable control measurements, mice received leptin infusion (2 μg·kg(-1)·day(-1) iv) for 7 days. Compared with Shp2(flox/flox) controls, Shp2/Pomc-cre mice at 22 wk of age were slightly heavier (34 ± 1 vs. 31 ± 1 g) but consumed a similar amount of food (3.9 ± 0.3 vs. 3.8 ± 0.2 g/day). Leptin infusion reduced food intake in Shp2(flox/flox) mice (2.6 ± 0.5 g) and Shp2/Pomc-cre mice (3.2 ± 0.3 g). Despite decreasing food intake, leptin infusion increased MAP in control mice, whereas no significant change in MAP was observed in Shp2/Pomc-cre mice. Leptin infusion also decreased plasma glucose and insulin levels in controls (12 ± 1 to 6 ± 1 μU/ml and 142 ± 12 to 81 ± 8 mg/100 ml) but not in Shp2/Pomc-cre mice. Leptin increased V̇o2 by 16 ± 2% in controls and 7 ± 1% in Shp2/Pomc-cre mice. These results indicate that Shp2 signaling in POMC neurons contributes to the long-term BP and antidiabetic actions of leptin and may play a modest role in normal regulation of body weight.
Collapse
Affiliation(s)
- Jussara M do Carmo
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, and Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi;
| | - Alexandre A da Silva
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, and Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sabira E Ebaady
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, and Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Price O Sessums
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, and Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ralph S Abraham
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, and Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| | - Joel K Elmquist
- Department of Pharmacology and Division of Hypothalamic Research, Department of Internal Medicine, Utah Southwestern Medical Center, Dallas, Texas; and
| | - Bradford B Lowell
- Division of Endocrinology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - John E Hall
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, and Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
22
|
Tajan M, Batut A, Cadoudal T, Deleruyelle S, Le Gonidec S, Saint Laurent C, Vomscheid M, Wanecq E, Tréguer K, De Rocca Serra-Nédélec A, Vinel C, Marques MA, Pozzo J, Kunduzova O, Salles JP, Tauber M, Raynal P, Cavé H, Edouard T, Valet P, Yart A. LEOPARD syndrome-associated SHP2 mutation confers leanness and protection from diet-induced obesity. Proc Natl Acad Sci U S A 2014; 111:E4494-503. [PMID: 25288766 PMCID: PMC4210352 DOI: 10.1073/pnas.1406107111] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
LEOPARD syndrome (multiple Lentigines, Electrocardiographic conduction abnormalities, Ocular hypertelorism, Pulmonary stenosis, Abnormal genitalia, Retardation of growth, sensorineural Deafness; LS), also called Noonan syndrome with multiple lentigines (NSML), is a rare autosomal dominant disorder associating various developmental defects, notably cardiopathies, dysmorphism, and short stature. It is mainly caused by mutations of the PTPN11 gene that catalytically inactivate the tyrosine phosphatase SHP2 (Src-homology 2 domain-containing phosphatase 2). Besides its pleiotropic roles during development, SHP2 plays key functions in energetic metabolism regulation. However, the metabolic outcomes of LS mutations have never been examined. Therefore, we performed an extensive metabolic exploration of an original LS mouse model, expressing the T468M mutation of SHP2, frequently borne by LS patients. Our results reveal that, besides expected symptoms, LS animals display a strong reduction of adiposity and resistance to diet-induced obesity, associated with overall better metabolic profile. We provide evidence that LS mutant expression impairs adipogenesis, triggers energy expenditure, and enhances insulin signaling, three features that can contribute to the lean phenotype of LS mice. Interestingly, chronic treatment of LS mice with low doses of MEK inhibitor, but not rapamycin, resulted in weight and adiposity gains. Importantly, preliminary data in a French cohort of LS patients suggests that most of them have lower-than-average body mass index, associated, for tested patients, with reduced adiposity. Altogether, these findings unravel previously unidentified characteristics for LS, which could represent a metabolic benefit for patients, but may also participate to the development or worsening of some traits of the disease. Beyond LS, they also highlight a protective role of SHP2 global LS-mimicking modulation toward the development of obesity and associated disorders.
Collapse
Affiliation(s)
- Mylène Tajan
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Aurélie Batut
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Thomas Cadoudal
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Simon Deleruyelle
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Sophie Le Gonidec
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Céline Saint Laurent
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Maëlle Vomscheid
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Estelle Wanecq
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Karine Tréguer
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Audrey De Rocca Serra-Nédélec
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Claire Vinel
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Marie-Adeline Marques
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Joffrey Pozzo
- Cardiology Unit, University Hospital Center of Rangueil Toulouse, F-31432 Toulouse, France
| | - Oksana Kunduzova
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Jean-Pierre Salles
- Endocrine, Bone Diseases, and Genetics Unit, Children's Hospital, University Hospital Center of Purpan Toulouse, F-31024 Toulouse, France
| | - Maithé Tauber
- Endocrine, Bone Diseases, and Genetics Unit, Children's Hospital, University Hospital Center of Purpan Toulouse, F-31024 Toulouse, France
| | - Patrick Raynal
- EA4568 Laboratoire Mécanismes des Cardiopathies et Résistances Hormonales dans le Syndrome de Noonan et les Syndromes Apparentés, Université de Toulouse, Université Paul Sabatier, F-31062 Toulouse, France; and
| | - Hélène Cavé
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche S1131, Unité de Formation et de Recherche de Médecine Paris-Diderot-Institut Universitaire d'Hématologie Département de Génétique, Unité Fonctionnelle de Génétique Moléculaire Hôpital Robert Debré, F-75019 Paris, France
| | - Thomas Edouard
- Endocrine, Bone Diseases, and Genetics Unit, Children's Hospital, University Hospital Center of Purpan Toulouse, F-31024 Toulouse, France
| | - Philippe Valet
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France
| | - Armelle Yart
- Institut National de la Santé et de la Recherche Médicale, U1048, F-31432 Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, Université Paul Sabatier, F-31432 Toulouse, France;
| |
Collapse
|
23
|
Knobler H, Elson A. Metabolic regulation by protein tyrosine phosphatases. J Biomed Res 2014; 28:157-68. [PMID: 25013399 PMCID: PMC4085553 DOI: 10.7555/jbr.28.20140012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 01/28/2014] [Indexed: 01/14/2023] Open
Abstract
Obesity and the metabolic syndrome and their associated morbidities are major public health issues, whose prevalence will continue to increase in the foreseeable future. Aberrant signaling by the receptors for leptin and insulin plays a pivotal role in development of the metabolic syndrome. More complete molecular-level understanding of how both of these key signaling pathways are regulated is essential for full characterization of obesity, the metabolic syndrome, and type II diabetes, and for developing novel treatments for these diseases. Phosphorylation of proteins on tyrosine residues plays a key role in mediating the effects of leptin and insulin on their target cells. Here, we discuss the molecular methods by which protein tyrosine phosphatases, which are key physiological regulators of protein phosphorylation in vivo, affect signaling by the leptin and insulin receptors in their major target tissues.
Collapse
Affiliation(s)
- Hilla Knobler
- Diabetes and Metabolic Disease Unit, Kaplan Medical Center, Rehovot 76100, Israel
| | - Ari Elson
- Department of Molecular Genetics, the Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
24
|
Vignozzi L, Filippi S, Comeglio P, Cellai I, Morelli A, Maneschi E, Sarchielli E, Gacci M, Carini M, Vannelli GB, Maggi M. Tadalafil effect on metabolic syndrome-associated bladder alterations: an experimental study in a rabbit model. J Sex Med 2014; 11:1159-72. [PMID: 24612540 DOI: 10.1111/jsm.12478] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Metabolic syndrome (MetS) and lower urinary tract symptoms (LUTS) are often associated. Bladder detrusor hyper-contractility-a major LUTS determinant-is characterized by increased Ras homolog gene family, member A/Rho-associated protein kinase (RhoA/ROCK) signaling, which is often upregulated in MetS. AIM This study investigated the effects of tadalafil dosing on RhoA/ROCK signaling in bladder, in a rabbit model of high-fat diet (HFD)-induced MetS. METHODS Adult male rabbits feeding a HFD for 12 weeks. A subset of HFD animals was treated with tadalafil (2 mg/kg/day, 1 week: the last of the 12 weeks) and compared with HFD and control (feeding a regular diet) rabbits. MAIN OUTCOME MEASURES In vitro contractility studies to evaluate the relaxant effect of the selective ROCK inhibitor, Y-27632, in carbachol precontracted bladder strips. Evaluation of RhoA activation by its membrane translocation. Immunohistochemistry for ROCK expression has been performed to evaluate ROCK expression in bladder from the different experimental groups. mRNA expression of inflammation, pro-fibrotic markers by quantitative RT-PCR has been performed to evaluate the effect of tadalafil on MetS-induced inflammation and fibrosis within the bladder. The in vitro effect of tadalafil on RhoA/ROCK signaling in bladder smooth muscle cells was evaluated by using chemotaxis assay. RESULTS Bladder strips from HFD rabbits showed hyper-responsiveness to Y-27632, indicating RhoA/ROCK overactivity in HFD bladder compared with matched controls. Accordingly, the fraction of activated (translocated to the membrane) RhoA as well as ROCK expression are increased in HFD bladder. Tadalafil dosing normalized HFD-induced bladder hypersensitivity to Y-27632, by reducing RhoA membrane translocation and ROCK overexpression. Tadalafil dosing reduced mRNA expression of inflammatory, pro-fibrotic, and hypoxia markers. A direct inhibitory effect of tadalafil on RhoA/ROCK signaling in bladder smooth muscle cell was demonstrated by using chemotaxis assay. Pre-treatment with tadalafil inhibited both basal and PDGF-induced migration of bladder smooth muscle cells. CONCLUSIONS Tadalafil dosing reduced RhoA/ROCK signaling and smooth muscle overactivity in an animal model of MetS-associated bladder alterations. Our findings suggest a novel mechanism of action of tadalafil in alleviating LUTS in MetS patients.
Collapse
Affiliation(s)
- Linda Vignozzi
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Almahariq M, Mei FC, Cheng X. Cyclic AMP sensor EPAC proteins and energy homeostasis. Trends Endocrinol Metab 2014; 25:60-71. [PMID: 24231725 PMCID: PMC3946731 DOI: 10.1016/j.tem.2013.10.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 09/30/2013] [Accepted: 10/11/2013] [Indexed: 12/16/2022]
Abstract
The pleiotropic second-messenger cAMP plays a crucial role in mediating the effects of various hormones on metabolism. The major intracellular functions of cAMP are transduced by protein kinase A (PKA) and by exchange proteins directly activated by cAMP (EPACs). The latter act as guanine-nucleotide exchange factors for the RAS-like small G proteins Rap1 and Rap2. Although the role of PKA in regulating energy balance has been extensively studied, the impact of EPACs remains relatively enigmatic. This review summarizes recent genetic and pharmacological studies concerning EPAC involvement in glucose homeostasis and energy balance via the regulation of leptin and insulin signaling pathways. In addition, the development of small-molecule EPAC-specific modulators and their therapeutic potential for the treatment of diabetes and obesity are discussed.
Collapse
Affiliation(s)
- Muayad Almahariq
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas 77555-0615, USA
| | - Fang C Mei
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas 77555-0615, USA; Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Xiaodong Cheng
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, Texas 77555-0615, USA; Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, Texas 77030, USA.
| |
Collapse
|
26
|
Comeglio P, Morelli A, Cellai I, Vignozzi L, Sarchielli E, Filippi S, Maneschi E, Corcetto F, Corno C, Gacci M, Vannelli GB, Maggi M. Opposite effects of tamoxifen on metabolic syndrome-induced bladder and prostate alterations: a role for GPR30/GPER? Prostate 2014; 74:10-28. [PMID: 24037776 DOI: 10.1002/pros.22723] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 07/23/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND BPH and LUTS have been associated to obesity, hypogonadism, and metabolic syndrome (MetS). MetS-induced prostate and bladder alterations, including inflammation and tissue remodeling, have been related to a low-testosterone and high-estrogen milieu. In addition to ERs, GPR30/GPER is able to mediate several estrogenic non-genomic actions. METHODS Supplementing a subgroup of MetS rabbits with tamoxifen, we analyzed the in vivo effects on MetS-induced prostate and bladder alterations. The effects of selective ER/GPER ligands and GPER silencing on prostate inflammation were also studied in vitro using hBPH cells. RESULTS ERα, ERβ, and PR expression was upregulated in MetS bladder, where tamoxifen decreased ERα and PR expression, further stimulating ERβ. In addition, tamoxifen-dosing decreased MetS-induced overexpression of inflammatory and tissue remodeling genes. In prostate, sex steroid receptors, pro-inflammatory and pro-fibrotic genes were upregulated in MetS. However, tamoxifen did not affect them and even increased COX-2. In hBPH cells, 17β-estradiol increased IL-8 secretion, an effect blunted by co-treatment with GPER antagonist G15 but not by ER antagonist ICI 182,780, which further increased it. GPER agonist G1 dose-dependently (IC50 = 1.6 nM) induced IL-8 secretion. In vitro analysis demonstrated that GPER silencing reverted these stimulatory effects. CONCLUSIONS GPER can be considered the main mediator of estrogen action in prostate, whereas in bladder the mechanism appears to rely on ERα, as indicated by in vivo experiments with tamoxifen dosing. Limiting the effects of the MetS-induced estrogen action via GPER could offer new perspectives in the management of BPH/LUTS, whereas tamoxifen dosing showed potential benefits in bladder.
Collapse
Affiliation(s)
- P Comeglio
- Sexual Medicine and Andrology Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
do Carmo JM, da Silva AA, Sessums PO, Ebaady SH, Pace BR, Rushing JS, Davis MT, Hall JE. Role of Shp2 in forebrain neurons in regulating metabolic and cardiovascular functions and responses to leptin. Int J Obes (Lond) 2013; 38:775-83. [PMID: 24030516 PMCID: PMC3954949 DOI: 10.1038/ijo.2013.177] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 08/29/2013] [Accepted: 09/05/2013] [Indexed: 02/04/2023]
Abstract
Objective We examined whether deficiency of Shp2 signaling in forebrain neurons alters metabolic and cardiovascular regulation under various conditions and if it attenuates the anorexic and cardiovascular effects of leptin. We also tested whether forebrain Shp2 deficiency alters blood pressure (BP) and heart rate (HR) responses to acute stress. Design Forebrain Shp2-/- mice were generated by crossing Shp2flox/flox mice with CamKIIα-cre mice. At 22 to 24 weeks of age, mice were instrumented for telemetry for measurement of BP, HR and body temperature (BT). Oxygen consumption (VO2), energy expenditure and motor activity were monitored by indirect calorimetry. Results Shp2/CamKIIα-cre mice were heavier (46±3 vs 32±1 g), hyperglycemic, hyperleptinemic, hyperinsulinemic, and hyperphagic compared to Shp2flox/flox control mice. Shp2/CamKIIα-cre mice exhibited reduced food intake responses to fasting/refeeding and impaired regulation of BT when exposed to 15°C and 30°C ambient temperatures. Despite being obese and having many features of metabolic syndrome, Shp2/CamKIIα-cre mice had similar daily average BP and HR compared to Shp2flox/flox mice (112±2 vs 113±1 mmHg and 595±34 vs 650±40 bpm), but exhibited increased BP and HR responses to cold exposure and acute air-jet stress test. Leptin's ability to reduce food intake and to raise BP were markedly attenuated in Shp2/CamKIIα-cre mice. Conclusion These results suggest that forebrain Shp2 signaling regulates food intake, appetite responses to caloric deprivation, and thermogenic control of body temperature during variations in ambient temperature. Deficiency of Shp2 signaling in the forebrain is associated with augmented cardiovascular responses to cold and acute stress but attenuated BP responses to leptin.
Collapse
Affiliation(s)
- J M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - A A da Silva
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - P O Sessums
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - S H Ebaady
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - B R Pace
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - J S Rushing
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - M T Davis
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - J E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
28
|
do Carmo JM, da Silva AA, Dubinion J, Sessums PO, Ebaady SH, Wang Z, Hall JE. Control of metabolic and cardiovascular function by the leptin-brain melanocortin pathway. IUBMB Life 2013; 65:692-8. [PMID: 23847053 DOI: 10.1002/iub.1187] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 05/11/2013] [Indexed: 12/26/2022]
Abstract
Obesity is recognized as a major worldwide health problem. Excess weight gain is the most common cause of elevated blood pressure (BP) and markedly increases the risk of metabolic, cardiovascular and renal diseases. Although the mechanisms linking obesity with hypertension have not been fully elucidated, increased sympathetic nervous system (SNS) activity contributes to elevated BP in obese subjects. Recent evidence indicates that leptin and the central nervous system (CNS) melanocortin system, including melanocortin 4 receptors (MC4R), play a key role in linking obesity with increased SNS activity and hypertension. Leptin, a peptide-hormone produced by adipose tissue, crosses the blood-brain barrier and activates brain centers that control multiple metabolic functions as well as SNS activity and BP via the CNS melanocortin system. The crosstalk between peripheral signals (e.g., leptin) and activation of CNS pathways (e.g., MC4R) that regulate energy balance, SNS activity and BP represents an important target for treating obesity and its metabolic and cardiovascular consequences.
Collapse
Affiliation(s)
- Jussara M do Carmo
- Department of Physiology and Biophysics, Center of Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Vignozzi L, Gacci M, Cellai I, Santi R, Corona G, Morelli A, Rastrelli G, Comeglio P, Sebastanelli A, Maneschi E, Nesi G, De Nunzio C, Tubaro A, Mannucci E, Carini M, Maggi M. Fat boosts, while androgen receptor activation counteracts, BPH-associated prostate inflammation. Prostate 2013; 73:789-800. [PMID: 23620238 DOI: 10.1002/pros.22623] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 11/02/2012] [Indexed: 01/04/2023]
Abstract
BACKGROUND Metabolic syndrome (MetS) and benign prostate hyperplasia (BPH) are often comorbid. Chronic inflammation, a determinant pathogenic factor for BPH, is a putative link between the two conditions. METHODS In a multi-center cohort of BPH patients (n = 244) who underwent prostatectomy, we evaluated whether MetS is associated with prostatic inflammation in BPH specimens. In addition, we investigated the in vitro inflammatory effects of metabolic insults on human prostatic myofibroblastic cells (hBPH). RESULTS Inflammatory infiltrates score (IS) in prostatectomy specimens showed a step-wise association with the number of MetS factors present (P = 0.001). After adjusting for age, reduced HDL cholesterol, and elevated triglycerides were the only factors significantly associated with IS. Increased IS was also significantly associated with hypogonadism. In an age- and testosterone (T)-adjusted model, dyslipidemia was still associated with IS. To investigate whether metabolic factors could directly trigger prostate inflammation, we performed preliminary studies in myofibroblastic hBPH. Among the different factors, oxidized low-density lipoprotein (oxLDL) showed the highest secretion of IL-8 (>10-fold)-a surrogate marker of prostate inflammation--as well as IL-6, and bFGF. Co-treatment with DHT significantly inhibited oxLDL-induced secretion of IL-8, whilst an AR-antagonist, bicalutamide, reversed DHT effects. DHT suppresses oxLDL receptor (LOX-1) expression. CONCLUSIONS Our data suggest that fats and insulin could have a detrimental effect on prostate health, boosting inflammation, a key pathogenic factor in BPH. Conversely, beneficial effects of DHT in counteracting lipid- and insulin-induced prostatic alterations, suggest that T-via its conversion into DHT-may have unexpected beneficial effects on prostate health.
Collapse
Affiliation(s)
- Linda Vignozzi
- Sexual Medicine and Andrology Unit, Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Jia ZF, Cao XY, Cao DH, Kong F, Kharbuja P, Jiang J. Polymorphisms of PTPN11 gene could influence serum lipid levels in a sex-specific pattern. Lipids Health Dis 2013; 12:72. [PMID: 23672255 PMCID: PMC3685535 DOI: 10.1186/1476-511x-12-72] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 05/10/2013] [Indexed: 01/18/2023] Open
Abstract
Background Previous studies have reported that different genotypes of PTPN11 gene (protein tyrosine phosphatase, non-receptor 11) were associated with different levels of serum lipids. The aim of this study was to explore the relationship between single nucleotide polymorphisms (SNPs) of PTPN11 and serum lipids in Northeast Chinese. Methods A total of 1003 subjects, 584 males and 419 females, were included in the study and their serum lipids were determined. Five htSNPs (rs2301756, rs12423190, rs12229892, rs7958372 and rs4767860) of PTPN11 gene were genotyped using TaqMan assay method. Results All of the five SNPs were in Hardy-Weinberg equilibrium. The male subjects had higher triglyceride (TG), higher low-density lipoprotein cholesterol (LDL-C) and lower high-density lipoprotein cholesterol (HDL-C) level than females. In males, rs4767860 was found to be associated with serum TG and total cholesterol (TC) levels and rs12229892 was associated with TC level. However, these significant associations could not be observed in females. In females, rs2301756 was found to be associated with TG and rs7958372 was associated with LDL-C level. Haplotype analysis showed that the GCGTG haplotype was associated with slightly higher TG level and ATGCG with higher TC level. Conclusions SNPs of PTPN11 may play a role in serum lipids in a sex-specific pattern. However, more studies are needed to confirm the conclusion and explore the underlying mechanism.
Collapse
Affiliation(s)
- Zhi-Fang Jia
- Division of Clinical Epidemiology, First Hospital of Jilin University, Changchun 130021, China
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
Leptin is secreted into the bloodstream by adipocytes and is required for the maintenance of energy homeostasis and body weight. Leptin deficiency or genetic defects in the components of the leptin signaling pathways cause obesity. Leptin controls energy balance and body weight mainly through leptin receptor b (LEPRb)-expressing neurons in the brain, particularly in the hypothalamus. These LEPRb-expressing neurons function as the first-order neurons that project to the second-order neurons located within and outside the hypothalamus, forming a neural network that controls the energy homeostasis and body weight. Multiple factors, including inflammation and endoplasmic reticulum (ER) stress, contribute to leptin resistance. Leptin resistance is the key risk factor for obesity. This review is focused on recent advance about leptin action, leptin signaling, and leptin resistance.
Collapse
|
32
|
Abstract
The blood-testis barrier (BTB) is a large junctional complex composed of tight junctions, adherens junctions, and gap junctions between adjacent Sertoli cells in the seminiferous tubules of the testis. Maintenance of the BTB as well as the controlled disruption and reformation of the barrier is essential for spermatogenesis and male fertility. Tyrosine phosphorylation of BTB proteins is known to regulate the integrity of adherens and tight junctions found at the BTB. SHP2 is a nonreceptor protein tyrosine phosphatase (PTP) and a key regulator of growth factor-mediated tyrosine kinase signaling pathways. We found that SHP2 is localized to Sertoli-Sertoli cell junctions in rat testis. The overexpression of a constitutive active SHP2 mutant, SHP2 Q79R, up-regulated the BTB disruptor ERK1/2 via Src kinase in primary rat Sertoli cells in culture. Furthermore, focal adhesion kinase (FAK), which also supports BTB integrity, was found to interact with SHP2 and constitutive activation of SHP2 decreased FAK tyrosine phosphorylation. Expression of the SHP2 Q79R mutant in primary cultured Sertoli cells also resulted in the loss of tight junction and adherens junction integrity that corresponded with the disruption of the actin cytoskeleton and mislocalization of adherens junction and tight junction proteins N-cadherin, β-catenin, and ZO-1 away from the plasma membrane. These results suggest that SHP2 is a key regulator of BTB integrity and Sertoli cell support of spermatogenesis and fertility.
Collapse
Affiliation(s)
- Pawan Puri
- Center for Research in Reproductive Physiology, Department of Cell Biology and Molecular Physiology, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | |
Collapse
|
33
|
Tsou RC, Bence KK. Central regulation of metabolism by protein tyrosine phosphatases. Front Neurosci 2013; 6:192. [PMID: 23308070 PMCID: PMC3538333 DOI: 10.3389/fnins.2012.00192] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 12/17/2012] [Indexed: 11/13/2022] Open
Abstract
Protein tyrosine phosphatases (PTPs) are important regulators of intracellular signaling pathways via the dephosphorylation of phosphotyrosyl residues on various receptor and non-receptor substrates. The phosphorylation state of central nervous system (CNS) signaling components underlies the molecular mechanisms of a variety of physiological functions including the control of energy balance and glucose homeostasis. In this review, we summarize the current evidence implicating PTPs as central regulators of metabolism, specifically highlighting their interactions with the neuronal leptin and insulin signaling pathways. We discuss the role of a number of PTPs (PTP1B, SHP2, TCPTP, RPTPe, and PTEN), reviewing the findings from genetic mouse models and in vitro studies which highlight these phosphatases as key central regulators of energy homeostasis.
Collapse
Affiliation(s)
- Ryan C Tsou
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania Philadelphia, PA, USA
| | | |
Collapse
|
34
|
He Z, Zhu HH, Bauler TJ, Wang J, Ciaraldi T, Alderson N, Li S, Raquil MA, Ji K, Wang S, Shao J, Henry RR, King PD, Feng GS. Nonreceptor tyrosine phosphatase Shp2 promotes adipogenesis through inhibition of p38 MAP kinase. Proc Natl Acad Sci U S A 2013; 110:E79-88. [PMID: 23236157 PMCID: PMC3538237 DOI: 10.1073/pnas.1213000110] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The molecular mechanism underlying adipogenesis and the physiological functions of adipose tissue are not fully understood. We describe here a unique mouse model of severe lipodystrophy. Ablation of Ptpn11/Shp2 in adipocytes, mediated by aP2-Cre, led to premature death, lack of white fat, low blood pressure, compensatory erythrocytosis, and hepatic steatosis in Shp2(fat-/-) mice. Fat transplantation partially rescued the lifespan and blood pressure in Shp2(fat-/-) mice, and administration of leptin also restored partially the blood pressure of mutant animals with endogenous leptin deficiency. Consistently, homozygous deletion of Shp2 inhibited adipocyte differentiation from embryonic stem (ES) cells. Biochemical analyses suggest a Shp2-TAO2-p38-p300-PPARγ pathway in adipogenesis, in which Shp2 suppresses p38 activation, leading to stabilization of p300 and enhanced PPARγ expression. Inhibition of p38 restored adipocyte differentiation from Shp2(-/-) ES cells, and p38 signaling is also suppressed in obese patients and obese animals. These results illustrate an essential role of adipose tissue in mammalian survival and physiology and also suggest a common signaling mechanism involved in adipogenesis and obesity development.
Collapse
Affiliation(s)
- Zhao He
- Department of Pathology and Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0864
| | - Helen H. Zhu
- Department of Pathology and Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0864
| | - Timothy J. Bauler
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109-5620; and
| | - Jing Wang
- Department of Pathology and Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0864
| | - Theodore Ciaraldi
- Veteran’s Administration San Diego Healthcare System and Department of Medicine, and
| | - Nazilla Alderson
- Department of Pathology and Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0864
| | - Shuangwei Li
- Department of Pathology and Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0864
| | - Marie-Astrid Raquil
- Department of Pathology and Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0864
| | - Kaihong Ji
- Department of Pathology and Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0864
| | - Shufen Wang
- Department of Pathology and Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0864
| | - Jianhua Shao
- Department of Pediatrics, University of California at San Diego, La Jolla,CA 92093
| | - Robert R. Henry
- Veteran’s Administration San Diego Healthcare System and Department of Medicine, and
| | - Philip D. King
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109-5620; and
| | - Gen-Sheng Feng
- Department of Pathology and Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0864
| |
Collapse
|
35
|
Enhanced leptin sensitivity, reduced adiposity, and improved glucose homeostasis in mice lacking exchange protein directly activated by cyclic AMP isoform 1. Mol Cell Biol 2012; 33:918-26. [PMID: 23263987 DOI: 10.1128/mcb.01227-12] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The prototypic second messenger cyclic AMP (cAMP) is essential for controlling cellular metabolism, including glucose and lipid homeostasis. In mammals, the majority of cAMP functions are mediated by cAMP-dependent protein kinase (PKA) and exchange proteins directly activated by cAMP (Epacs). To explore the physiological functions of Epac1, we generated Epac1 knockout mice. Here we report that Epac1 null mutants have reduced white adipose tissue and reduced plasma leptin levels but display heightened leptin sensitivity. Epac1-deficient mice are more resistant to high-fat diet-induced obesity, hyperleptinemia, and glucose intolerance. Furthermore, pharmacological inhibition of Epac by use of an Epac-specific inhibitor reduces plasma leptin levels in vivo and enhances leptin signaling in organotypic hypothalamic slices. Taken together, our results demonstrate that Epac1 plays an important role in regulating adiposity and energy balance.
Collapse
|
36
|
Novel role for SHP-2 in nutrient-responsive control of S6 kinase 1 signaling. Mol Cell Biol 2012; 33:293-306. [PMID: 23129808 DOI: 10.1128/mcb.01285-12] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Amino acids are required for the activation of the mammalian target of rapamycin complex 1 (mTORC1), which plays a critical role in cell growth, proliferation, and metabolism. The branched-chain amino acid leucine is an essential nutrient that stimulates mTORC1 to promote protein synthesis by activating p70 S6 kinase 1 (S6K1). Here we show that the protein tyrosine phosphatase SHP-2 is required for leucine-induced activation of S6K1 in skeletal myoblasts. In response to leucine, S6K1 activation is inhibited in myoblasts either lacking SHP-2 expression or overexpressing a catalytically inactive mutant of SHP-2. Activation of S6K1 by leucine requires the mobilization of intracellular calcium (Ca(2+)), which we show is mediated by SHP-2 in an inositol-1,4,5-trisphosphate-dependent manner. Ectopic Ca(2+) mobilization rescued the S6K1 activation defect in SHP-2-deficient myoblasts. SHP-2 was identified to act upstream of phospholipase C β4, linking it to the generation of nutrient-induced Ca(2+) release and S6K1 phosphorylation. Consistent with these results, SHP-2-deficient myoblasts exhibited impaired leucine sensing, leading to defective autophagy and reduced myoblast size. These data define a new role for SHP-2 as a nutrient-sensing regulator in skeletal myoblasts that is required for the activation of S6K1.
Collapse
|
37
|
Morelli A, Comeglio P, Filippi S, Sarchielli E, Cellai I, Vignozzi L, Yehiely-Cohen R, Maneschi E, Gacci M, Carini M, Adorini L, Vannelli GB, Maggi M. Testosterone and farnesoid X receptor agonist INT-747 counteract high fat diet-induced bladder alterations in a rabbit model of metabolic syndrome. J Steroid Biochem Mol Biol 2012; 132:80-92. [PMID: 22406511 DOI: 10.1016/j.jsbmb.2012.02.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 02/27/2012] [Accepted: 02/28/2012] [Indexed: 12/20/2022]
Abstract
In the male, metabolic syndrome (MetS) is associated to an increased risk of benign prostatic hyperplasia (BPH) and lower urinary tract symptoms (LUTS). A recently established rabbit model of high fat diet (HFD)-induced MetS showed hypogonadism and the presence of prostate gland alterations, including inflammation, hypoxia and fibrosis. The present study investigated whether HFD-induced MetS might also alter bladder structure and function. Testosterone and the farnesoid X receptor (FXR) agonist INT-747, were evaluated for possible effects on HFD bladder. MetS rabbits develop bladder alterations, including fibrosis (reduced muscle/fiber ratio), hypoxia [2-fold increase as compared to regular diet (RD) group], low-grade inflammation (increased leukocyte infiltration and inflammatory markers) and RhoA/ROCK hyperactivity. Bladder strips from HFD rabbits, pre-contracted with carbachol, showed an overactive response to the selective ROCK inhibitor Y-27632. All these HFD-induced bladder alterations were partially blunted by testosterone and almost completely reverted by INT-747. Both treatments prevented some MetS features (glucose intolerance and visceral fat increase), thus suggesting that their effects on bladder could be ascribed to an improvement of the metabolic and/or hypogonadal state. However, a pathogenetic role for hypogonadism has been ruled out as GnRH analog-induced hypogonadal rabbits, fed a regular diet, did not show any detectable bladder alterations. In addition, INT-747 did not revert the MetS-induced hypogonadal state. FXR mRNA was highly expressed in rabbit bladder and positively associated with visceral fat increase. A direct effect of INT-747 on bladder smooth muscle was further suggested by inhibition of RhoA/ROCK-mediated activity by in vitro experiments on isolated cells. In conclusion, HFD-related MetS features are associated to bladder derangements, which are ameliorated by testosterone or INT-747 administration. INT-747 showed the most marked effects in counteracting MetS-related RhoA/ROCK overactivity, thus opening novel therapeutic opportunities for this drug.
Collapse
Affiliation(s)
- Annamaria Morelli
- Department of Clinical Physiopathology, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Nagata N, Matsuo K, Bettaieb A, Bakke J, Matsuo I, Graham J, Xi Y, Liu S, Tomilov A, Tomilova N, Gray S, Jung DY, Ramsey JJ, Kim JK, Cortopassi G, Havel PJ, Haj FG. Hepatic Src homology phosphatase 2 regulates energy balance in mice. Endocrinology 2012; 153:3158-69. [PMID: 22619361 PMCID: PMC3380313 DOI: 10.1210/en.2012-1406] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The Src homology 2 domain-containing protein-tyrosine phosphatase Src homology phosphatase 2 (Shp2) is a negative regulator of hepatic insulin action in mice fed regular chow. To investigate the role of hepatic Shp2 in lipid metabolism and energy balance, we determined the metabolic effects of its deletion in mice challenged with a high-fat diet (HFD). We analyzed body mass, lipid metabolism, insulin sensitivity, and glucose tolerance in liver-specific Shp2-deficient mice (referred to herein as LSHKO) and control mice fed HFD. Hepatic Shp2 protein expression is regulated by nutritional status, increasing in mice fed HFD and decreasing during fasting. LSHKO mice gained less weight and exhibited increased energy expenditure compared with control mice. In addition, hepatic Shp2 deficiency led to decreased liver steatosis, enhanced insulin-induced suppression of hepatic glucose production, and impeded the development of insulin resistance after high-fat feeding. At the molecular level, LSHKO exhibited decreased hepatic endoplasmic reticulum stress and inflammation compared with control mice. In addition, tyrosine and serine phosphorylation of total and mitochondrial signal transducer and activator of transcription 3 were enhanced in LSHKO compared with control mice. In line with this observation and the increased energy expenditure of LSHKO, oxygen consumption rate was higher in liver mitochondria of LSHKO compared with controls. Collectively, these studies identify hepatic Shp2 as a novel regulator of systemic energy balance under conditions of high-fat feeding.
Collapse
Affiliation(s)
- Naoto Nagata
- Department of Nutrition, University of California Davis, Davis, California 95616, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sharma N, Kumar V, Everingham S, Mali RS, Kapur R, Zeng LF, Zhang ZY, Feng GS, Hartmann K, Roers A, Craig AWB. SH2 domain-containing phosphatase 2 is a critical regulator of connective tissue mast cell survival and homeostasis in mice. Mol Cell Biol 2012; 32:2653-63. [PMID: 22566685 PMCID: PMC3416204 DOI: 10.1128/mcb.00308-12] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 04/28/2012] [Indexed: 12/28/2022] Open
Abstract
Mast cells require KIT receptor tyrosine kinase signaling for development and survival. Here, we report that SH2 domain-containing phosphatase 2 (SHP2) signaling downstream of KIT is essential for mast cell survival and homeostasis in mice. Using a novel mouse model with shp2 deletion within mature mast cells (MC-shp2 knockout [KO]), we find that SHP2 is required for the homeostasis of connective tissue mast cells. Consistently with the loss of skin mast cells, MC-shp2 KO mice fail to mount a passive late-phase cutaneous anaphylaxis response. To better define the phenotype of shp2-deficient mast cells, we used an inducible shp2 knockout approach in bone marrow-derived mast cells (BMMCs) or cultured peritoneal mast cells and found that SHP2 promotes mast cell survival. We show that SHP2 promotes KIT signaling to extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase and downregulation of the proapoptotic protein Bim in BMMCs. Also, SHP2-deficient BMMCs failed to repopulate mast cells in mast cell-deficient mice. Silencing of Bim partially rescued survival defects in shp2-deficient BMMCs, consistent with the importance of a KIT → SHP2 → Ras/ERK pathway in suppressing Bim and promoting mast cell survival. Thus, SHP2 is a key node in a mast cell survival pathway and a new potential therapeutic target in diseases involving mast cells.
Collapse
Affiliation(s)
- Namit Sharma
- Department of Biomedical and Molecular Sciences, Division of Cancer Biology & Genetics, Queen's University, Kingston, Ontario, Canada
| | - Vijay Kumar
- Department of Biomedical and Molecular Sciences, Division of Cancer Biology & Genetics, Queen's University, Kingston, Ontario, Canada
| | - Stephanie Everingham
- Department of Biomedical and Molecular Sciences, Division of Cancer Biology & Genetics, Queen's University, Kingston, Ontario, Canada
| | - Raghuveer Singh Mali
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indianapolis, Indiana, USA
| | - Reuben Kapur
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indianapolis, Indiana, USA
| | - Li-Fan Zeng
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Bloomington, Indiana, USA
| | - Zhong-Yin Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Bloomington, Indiana, USA
| | - Gen-Sheng Feng
- Department of Pathology, University of California, San Diego, La Jolla, California, USA
| | - Karin Hartmann
- Department of Dermatology, University of Cologne, Cologne, Germany
| | - Axel Roers
- Institute for Immunology, Technical University of Dresden, Dresden, Germany
| | - Andrew W. B. Craig
- Department of Biomedical and Molecular Sciences, Division of Cancer Biology & Genetics, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
40
|
Vignozzi L, Cellai I, Santi R, Lombardelli L, Morelli A, Comeglio P, Filippi S, Logiodice F, Carini M, Nesi G, Gacci M, Piccinni MP, Adorini L, Maggi M. Antiinflammatory effect of androgen receptor activation in human benign prostatic hyperplasia cells. J Endocrinol 2012; 214:31-43. [PMID: 22562653 DOI: 10.1530/joe-12-0142] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Progression of benign prostatic hyperplasia (BPH) involves chronic inflammation and immune dysregulation. Preclinical studies have demonstrated that prostate inflammation and tissue remodeling are exacerbated by hypogonadism and prevented by testosterone supplementation. We now investigated whether, in humans, hypogonadism was associated with more severe BPH inflammation and the in vitro effect of the selective androgen receptor agonist dihydrotestosterone (DHT) on cultures of stromal cells derived from BPH patients (hBPH). Histological analysis of inflammatory infiltrates in prostatectomy specimens from a cohort of BPH patients and correlation with serum testosterone level was performed. Even after adjusting for confounding factors, hypogonadism was associated with a fivefold increased risk of intraprostatic inflammation, which was also more severe than that observed in eugonadal BPH patients. Triggering hBPH cells by inflammatory stimuli (tumor necrosis factor α, lipopolysaccharide, or CD4(+)T cells) induced abundant secretion of inflammatory/growth factors (interleukin 6 (IL6), IL8, and basic fibroblast growth factor (bFGF)). Co-culture of CD4(+)T cells with hBPH cells induced secretion of Th1 inducer (IL12), Th1-recruiting chemokine (interferon γ inducible protein 10, IP10), and Th2 (IL9)- and Th17 (IL17)-specific cytokines. Pretreatment with DHT inhibited NF-κB activation and suppressed secretion of several inflammatory/growth factors, with the most pronounced effects on IL8, IL6, and bFGF. Reduced inflammatory cytokine production by T-cells, an increase in IL10, and a significant reduction of T cells proliferation suggested that DHT exerted a broad anti inflammatory effect on testosterone cells [corrected]. In conclusion, our data demonstrate that DHT exerts an immune regulatory role on human prostatic stromal cells, inhibiting their potential to actively induce and/or sustain autoimmune and inflammatory responses.
Collapse
Affiliation(s)
- Linda Vignozzi
- Sexual Medicine and Andrology Unit, Department of Clinical Physiopathology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Shp2 controls female body weight and energy balance by integrating leptin and estrogen signals. Mol Cell Biol 2012; 32:1867-78. [PMID: 22431513 DOI: 10.1128/mcb.06712-11] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
In mammals, leptin regulates food intake and energy balance mainly through the activation of LepRb in the hypothalamus, and estrogen has a leptin-like effect in the hypothalamic control of metabolism. However, it remains to be elucidated how estrogen signaling is intertwined with the leptin pathway. We show here that Shp2, a nonreceptor tyrosine phosphatase, acts to integrate leptin and estrogen signals. The expression of a dominant-active mutant (Shp2(D61A)) in forebrain neurons conferred female, but not male, transgenic mice resistance to high-fat diet (HFD)-induced obesity and liver steatosis, accompanied by improved insulin sensitivity and glucose homeostasis. Fed with either HFD or regular chow food, Shp2(D61A) female mice showed dramatically enhanced leptin sensitivity. Microinjection of Shp2(D61A)-expressing adeno-associated virus into mediobasal hypothalamus elicited a similar antiobese effect in female mice. Biochemical analyses showed a physical association of Shp2 with estrogen receptor alpha, which is necessary for the synergistic and persistent activation of Erk by leptin and estrogen. Together, these results elucidate a mechanism for the direct cross talk of leptin and estrogen signaling and offer one explanation for the propensity of postmenopausal women to develop obesity.
Collapse
|
42
|
St-Pierre J, Tremblay ML. Modulation of leptin resistance by protein tyrosine phosphatases. Cell Metab 2012; 15:292-7. [PMID: 22405067 DOI: 10.1016/j.cmet.2012.02.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 01/31/2012] [Accepted: 02/06/2012] [Indexed: 10/28/2022]
Abstract
Leptin plays a central role in weight control by suppressing food intake and increasing energy expenditure. The concept of leptin resistance emerged to explain the seemingly paradoxical elevated leptin levels in obesity. Recent discoveries reveal that protein tyrosine phosphatases are key players in leptin resistance by globally suppressing leptin signaling.
Collapse
Affiliation(s)
- Julie St-Pierre
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec H3A 1A3, Canada.
| | | |
Collapse
|
43
|
Paternal age effect mutations and selfish spermatogonial selection: causes and consequences for human disease. Am J Hum Genet 2012; 90:175-200. [PMID: 22325359 DOI: 10.1016/j.ajhg.2011.12.017] [Citation(s) in RCA: 227] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 12/05/2011] [Accepted: 12/26/2011] [Indexed: 12/25/2022] Open
Abstract
Advanced paternal age has been associated with an increased risk for spontaneous congenital disorders and common complex diseases (such as some cancers, schizophrenia, and autism), but the mechanisms that mediate this effect have been poorly understood. A small group of disorders, including Apert syndrome (caused by FGFR2 mutations), achondroplasia, and thanatophoric dysplasia (FGFR3), and Costello syndrome (HRAS), which we collectively term "paternal age effect" (PAE) disorders, provides a good model to study the biological and molecular basis of this phenomenon. Recent evidence from direct quantification of PAE mutations in sperm and testes suggests that the common factor in the paternal age effect lies in the dysregulation of spermatogonial cell behavior, an effect mediated molecularly through the growth factor receptor-RAS signal transduction pathway. The data show that PAE mutations, although arising rarely, are positively selected and expand clonally in normal testes through a process akin to oncogenesis. This clonal expansion, which is likely to take place in the testes of all men, leads to the relative enrichment of mutant sperm over time-explaining the observed paternal age effect associated with these disorders-and in rare cases to the formation of testicular tumors. As regulation of RAS and other mediators of cellular proliferation and survival is important in many different biological contexts, for example during tumorigenesis, organ homeostasis and neurogenesis, the consequences of selfish mutations that hijack this process within the testis are likely to extend far beyond congenital skeletal disorders to include complex diseases, such as neurocognitive disorders and cancer predisposition.
Collapse
|
44
|
Vignozzi L, Morelli A, Sarchielli E, Comeglio P, Filippi S, Cellai I, Maneschi E, Serni S, Gacci M, Carini M, Piccinni MP, Saad F, Adorini L, Vannelli GB, Maggi M. Testosterone protects from metabolic syndrome-associated prostate inflammation: an experimental study in rabbit. J Endocrinol 2012; 212:71-84. [PMID: 22010203 DOI: 10.1530/joe-11-0289] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Metabolic syndrome (MetS) and benign prostatic hyperplasia (BPH)/lower urinary tract symptoms (LUTS) are often associated. One of their common denominators is hypogonadism. However, testosterone supplementation is limited by concerns for potential prostatic side effects. The objective was to determine whether MetS-associated prostate alterations are prevented by testosterone supplementation. We used a previously described animal model of MetS, obtained by feeding male rabbits a high-fat diet (HFD) for 12 weeks. Subsets of HFD rabbits were treated with testosterone or with the farnesoid X receptor agonist INT-747. Rabbits fed a standard diet were used as controls. HFD-animals develop hypogonadism and all the MetS features: hyperglycemia, glucose intolerance, dyslipidemia, hypertension, and visceral obesity. In addition, HFD-animals show a prostate inflammation. Immunohistochemical analysis demonstrated that HFD-induced prostate fibrosis, hypoxia, and inflammation. The mRNA expression of several proinflammatory (IL8, IL6, IL1β, and TNFα), T lymphocyte (CD4, CD8, Tbet, Gata3, and ROR γt), macrophage (TLR2, TLR4, and STAMP2), neutrophil (lactoferrin), inflammation (COX2 and RAGE), and fibrosis/myofibroblast activation (TGFβ, SM22α, αSMA, RhoA, and ROCK1/ROCK2) markers was significantly increased in HFD prostate. Testosterone, as well as INT-747, treatment prevented some MetS features, although only testosterone normalized all the HFD-induced prostate alterations. Interestingly, the ratio between testosterone and estradiol plasma level retains a significant, negative, association with all the fibrosis and the majority of inflammatory markers analyzed. These data highlight that testosterone protects rabbit prostate from MetS-induced prostatic hypoxia, fibrosis, and inflammation, which can play a role toward the development/progression of BPH/LUTS.
Collapse
Affiliation(s)
- Linda Vignozzi
- Sexual Medicine and Andrology Unit, Department of Clinical Physiopathology, University of Florence, Viale Pieraccini 6, Florence 50139, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Krajewska M, You Z, Rong J, Kress C, Huang X, Yang J, Kyoda T, Leyva R, Banares S, Hu Y, Sze CH, Whalen MJ, Salmena L, Hakem R, Head BP, Reed JC, Krajewski S. Neuronal deletion of caspase 8 protects against brain injury in mouse models of controlled cortical impact and kainic acid-induced excitotoxicity. PLoS One 2011; 6:e24341. [PMID: 21957448 PMCID: PMC3174961 DOI: 10.1371/journal.pone.0024341] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 08/09/2011] [Indexed: 11/25/2022] Open
Abstract
Background Acute brain injury is an important health problem. Given the critical position of caspase 8 at the crossroads of cell death pathways, we generated a new viable mouse line (Ncasp8−/−), in which the gene encoding caspase 8 was selectively deleted in neurons by cre-lox system. Methodology/Principal Findings Caspase 8 deletion reduced rates of neuronal cell death in primary neuronal cultures and in whole brain organotypic coronal slice cultures prepared from 4 and 8 month old mice and cultivated up to 14 days in vitro. Treatments of cultures with recombinant murine TNFα (100 ng/ml) or TRAIL (250 ng/mL) plus cyclohexamide significantly protected neurons against cell death induced by these apoptosis-inducing ligands. A protective role of caspase 8 deletion in vivo was also demonstrated using a controlled cortical impact (CCI) model of traumatic brain injury (TBI) and seizure-induced brain injury caused by kainic acid (KA). Morphometric analyses were performed using digital imaging in conjunction with image analysis algorithms. By employing virtual images of hundreds of brain sections, we were able to perform quantitative morphometry of histological and immunohistochemical staining data in an unbiased manner. In the TBI model, homozygous deletion of caspase 8 resulted in reduced lesion volumes, improved post-injury motor performance, superior learning and memory retention, decreased apoptosis, diminished proteolytic processing of caspases and caspase substrates, and less neuronal degeneration, compared to wild type, homozygous cre, and caspase 8-floxed control mice. In the KA model, Ncasp8−/− mice demonstrated superior survival, reduced seizure severity, less apoptosis, and reduced caspase 3 processing. Uninjured aged knockout mice showed improved learning and memory, implicating a possible role for caspase 8 in cognitive decline with aging. Conclusions Neuron-specific deletion of caspase 8 reduces brain damage and improves post-traumatic functional outcomes, suggesting an important role for this caspase in pathophysiology of acute brain trauma.
Collapse
Affiliation(s)
- Maryla Krajewska
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Zerong You
- Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Juan Rong
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Christina Kress
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Xianshu Huang
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Jinsheng Yang
- Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Tiffany Kyoda
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Ricardo Leyva
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Steven Banares
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Yue Hu
- Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
- VA San Diego Healthcare System, San Diego, California, United States of America
| | - Chia-Hung Sze
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Michael J. Whalen
- Neuroscience Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Leonardo Salmena
- Department of Medical Biophysics, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Razqallah Hakem
- Department of Medical Biophysics, Ontario Cancer Institute, Toronto, Ontario, Canada
| | - Brian P. Head
- Department of Anesthesiology, University of California San Diego, La Jolla, California, United States of America
| | - John C. Reed
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
- * E-mail: (SK); (JCR)
| | - Stan Krajewski
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
- * E-mail: (SK); (JCR)
| |
Collapse
|
46
|
Cooke M, Orlando U, Maloberti P, Podestá EJ, Cornejo Maciel F. Tyrosine phosphatase SHP2 regulates the expression of acyl-CoA synthetase ACSL4. J Lipid Res 2011; 52:1936-48. [PMID: 21903867 DOI: 10.1194/jlr.m015552] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Acyl-CoA synthetase 4 (ACSL4) is implicated in fatty acid metabolism with marked preference for arachidonic acid (AA). ACSL4 plays crucial roles in physiological functions such as steroid synthesis and in pathological processes such as tumorigenesis. However, factors regulating ACSL4 mRNA and/or protein levels are not fully described. Because ACSL4 protein expression requires tyrosine phosphatase activity, in this study we aimed to identify the tyrosine phosphatase involved in ACSL4 expression. NSC87877, a specific inhibitor of the tyrosine phosphatase SHP2, reduced ACSL4 protein levels in ACSL4-rich breast cancer cells and steroidogenic cells. Indeed, overexpression of an active form of SHP2 increased ACSL4 protein levels in MA-10 Leydig steroidogenic cells. SHP2 has to be activated through a cAMP-dependent pathway to exert its effect on ACSL4. The effects could be specifically attributed to SHP2 because knockdown of the phosphatase reduced ACSL4 mRNA and protein levels. Through the action on ACSL4 protein levels, SHP2 affected AA-CoA production and metabolism and, finally, the steroidogenic capacity of MA-10 cells: overexpression (or knockdown) of SHP2 led to increased (or decreased) steroid production. We describe for the first time the involvement of SHP2 activity in the regulation of the expression of the fatty acid-metabolizing enzyme ACSL4.
Collapse
Affiliation(s)
- Mariana Cooke
- Department of Biochemistry, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
47
|
Rousso-Noori L, Knobler H, Levy-Apter E, Kuperman Y, Neufeld-Cohen A, Keshet Y, Akepati VR, Klinghoffer RA, Chen A, Elson A. Protein tyrosine phosphatase epsilon affects body weight by downregulating leptin signaling in a phosphorylation-dependent manner. Cell Metab 2011; 13:562-72. [PMID: 21531338 DOI: 10.1016/j.cmet.2011.02.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 12/28/2010] [Accepted: 02/18/2011] [Indexed: 01/17/2023]
Abstract
Molecular-level understanding of body weight control is essential for combating obesity. We show that female mice lacking tyrosine phosphatase epsilon (RPTPe) are protected from weight gain induced by high-fat food, ovariectomy, or old age and exhibit increased whole-body energy expenditure and decreased adiposity. RPTPe-deficient mice, in particular males, exhibit improved glucose homeostasis. Female nonobese RPTPe-deficient mice are leptin hypersensitive and exhibit reduced circulating leptin concentrations, suggesting that RPTPe inhibits hypothalamic leptin signaling in vivo. Leptin hypersensitivity persists in aged, ovariectomized, and high-fat-fed RPTPe-deficient mice, indicating that RPTPe helps establish obesity-associated leptin resistance. RPTPe associates with and dephosphorylates JAK2, thereby downregulating leptin receptor signaling. Leptin stimulation induces phosphorylation of hypothalamic RPTPe at its C-terminal Y695, which drives RPTPe to downregulate JAK2. RPTPe is therefore an inhibitor of hypothalamic leptin signaling in vivo, and provides controlled negative-feedback regulation of this pathway following its activation.
Collapse
Affiliation(s)
- Liat Rousso-Noori
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bu Y, Shi T, Meng M, Kong G, Tian Y, Chen Q, Yao X, Feng G, Chen H, Cheng H, Lu Z. A novel screening model for the molecular drug for diabetes and obesity based on tyrosine phosphatase Shp2. Bioorg Med Chem Lett 2010; 21:874-8. [PMID: 21169016 DOI: 10.1016/j.bmcl.2010.11.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 11/06/2010] [Accepted: 11/09/2010] [Indexed: 10/18/2022]
Abstract
Tyrosine phosphatase Src-homology phosphotyrosyl phosphatase 2 (Shp2) was identified as a potential molecular target for therapeutic treatment of diabetes and obesity. However, there is still no systematic research on the enhancers for the Shp2 enzyme. The present study established a novel powerful model for the high-throughput screening of Shp2 enhancers and successfully identified a new specific Shp2 enhancer, oleanolic acid, from Chinese herbs.
Collapse
Affiliation(s)
- Yanyan Bu
- Xiamen City Key Laboratory of Metabolism Disease and Metabolic Disease Research Center, Institute for Biomedical Research, Lu Jiaxi Hall, Room 630, Xiamen University, Xiamen, Fujian 361005, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Bauler TJ, Kamiya N, Lapinski PE, Langewisch E, Mishina Y, Wilkinson JE, Feng GS, King PD. Development of severe skeletal defects in induced SHP-2-deficient adult mice: a model of skeletal malformation in humans with SHP-2 mutations. Dis Model Mech 2010; 4:228-39. [PMID: 21068439 PMCID: PMC3046097 DOI: 10.1242/dmm.006130] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
SHP-2 (encoded by PTPN11) is a ubiquitously expressed protein tyrosine phosphatase required for signal transduction by multiple different cell surface receptors. Humans with germline SHP-2 mutations develop Noonan syndrome or LEOPARD syndrome, which are characterized by cardiovascular, neurological and skeletal abnormalities. To study how SHP-2 regulates tissue homeostasis in normal adults, we used a conditional SHP-2 mouse mutant in which loss of expression of SHP-2 was induced in multiple tissues in response to drug administration. Induced deletion of SHP-2 resulted in impaired hematopoiesis, weight loss and lethality. Most strikingly, induced SHP-2-deficient mice developed severe skeletal abnormalities, including kyphoses and scolioses of the spine. Skeletal malformations were associated with alterations in cartilage and a marked increase in trabecular bone mass. Osteoclasts were essentially absent from the bones of SHP-2-deficient mice, thus accounting for the osteopetrotic phenotype. Studies in vitro revealed that osteoclastogenesis that was stimulated by macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor kappa B ligand (RANKL) was defective in SHP-2-deficient mice. At least in part, this was explained by a requirement for SHP-2 in M-CSF-induced activation of the pro-survival protein kinase AKT in hematopoietic precursor cells. These findings illustrate an essential role for SHP-2 in skeletal growth and remodeling in adults, and reveal some of the cellular and molecular mechanisms involved. The model is predicted to be of further use in understanding how SHP-2 regulates skeletal morphogenesis, which could lead to the development of novel therapies for the treatment of skeletal malformations in human patients with SHP-2 mutations.
Collapse
Affiliation(s)
- Timothy J Bauler
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109-5620, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Deregulation of signaling pathways, through mutation or other molecular changes, can ultimately result in disease. The tyrosine phosphatase Shp2 has emerged as a major regulator of receptor tyrosine kinase (RTK) and cytokine receptor signaling. In the last decade, germline mutations in the human PTPN11 gene, encoding Shp2, were linked to Noonan (NS) and LEOPARD syndromes, two multisymptomatic developmental disorders that are characterized by short stature, craniofacial defects, cardiac defects, and mental retardation. Somatic Shp2 mutations are also associated with several types of human malignancies, such as the most common juvenile leukemia, juvenile myelomonocytic leukemia (JMML). Whereas NS and JMML are caused by gain-of-function (GOF) mutations of Shp2, loss-of-function (LOF) mutations are thought to be associated with LEOPARD syndrome. Animal models that carry conditional LOF and GOF mutations have allowed a better understanding of the mechanism of Shp2 function in disease, and shed light on the role of Shp2 in signaling pathways that control decisive events during embryonic development or during cellular transformation/tumorigenesis.
Collapse
|