101
|
Brown JA, Bugescu R, Mayer TA, Gata-Garcia A, Kurt G, Woodworth HL, Leinninger GM. Loss of Action via Neurotensin-Leptin Receptor Neurons Disrupts Leptin and Ghrelin-Mediated Control of Energy Balance. Endocrinology 2017; 158:1271-1288. [PMID: 28323938 PMCID: PMC5460836 DOI: 10.1210/en.2017-00122] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/06/2017] [Indexed: 01/30/2023]
Abstract
The hormones ghrelin and leptin act via the lateral hypothalamic area (LHA) to modify energy balance, but the underlying neural mechanisms remain unclear. We investigated how leptin and ghrelin engage LHA neurons to modify energy balance behaviors and whether there is any crosstalk between leptin and ghrelin-responsive circuits. We demonstrate that ghrelin activates LHA neurons expressing hypocretin/orexin (OX) to increase food intake. Leptin mediates anorectic actions via separate neurons expressing the long form of the leptin receptor (LepRb), many of which coexpress the neuropeptide neurotensin (Nts); we refer to these as NtsLepRb neurons. Because NtsLepRb neurons inhibit OX neurons, we hypothesized that disruption of the NtsLepRb neuronal circuit would impair both NtsLepRb and OX neurons from responding to their respective hormonal cues, thus compromising adaptive energy balance. Indeed, mice with developmental deletion of LepRb specifically from NtsLepRb neurons exhibit blunted adaptive responses to leptin and ghrelin that discoordinate the mesolimbic dopamine system and ingestive and locomotor behaviors, leading to weight gain. Collectively, these data reveal a crucial role for LepRb in the proper formation of LHA circuits, and that NtsLepRb neurons are important neuronal hubs within the LHA for hormone-mediated control of ingestive and locomotor behaviors.
Collapse
Affiliation(s)
- Juliette A. Brown
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
| | - Raluca Bugescu
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824
| | - Thomas A. Mayer
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824
| | - Adriana Gata-Garcia
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109
| | - Gizem Kurt
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824
| | - Hillary L. Woodworth
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824
| | - Gina M. Leinninger
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan 48824
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824
| |
Collapse
|
102
|
Uldall M, Bhatt DK, Kruuse C, Juhler M, Jansen-Olesen I, Jensen RH. Choroid plexus aquaporin 1 and intracranial pressure are increased in obese rats: towards an idiopathic intracranial hypertension model? Int J Obes (Lond) 2017; 41:1141-1147. [PMID: 28344346 DOI: 10.1038/ijo.2017.83] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/03/2017] [Accepted: 03/06/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND/OBJECTIVES Idiopathic intracranial hypertension (IIH) is a condition of increased intracranial pressure (ICP) without identifiable cause. The majority of IIH patients are obese, which suggests a connection between ICP and obesity. The aim of the study was to compare ICP in lean and obese rats. We also aimed to clarify if any ICP difference could be attributed to changes in some well-known ICP modulators; retinol and arterial partial pressure of CO2 (pCO2). Another potential explanation could be differences in water transport across the choroid plexus (CP) epithelia, and thus we furthermore investigated expression profiles of aquaporin 1 (AQP1) and Na/K ATPase. METHODS ICP was measured in obese and lean Zucker rats over a period of 28 days. Arterial pCO2 and serum retinol were measured in serum samples. The CPs were isolated, and target messenger RNA (mRNA) and protein were analyzed by quantitative PCR and western blot, respectively. RESULTS Obese rats had elevated ICP compared to lean controls on all recording days except day 0 (P<0.001). Serum retinol (P=0.35) and arterial pCO2 (P=0.16) did not differ between the two groups. Both AQP1 mRNA and protein levels were increased in the CP of the obese rats compared to lean rats (P=0.0422 and P=0.0281). There was no difference in Na/K ATPase mRNA or protein levels (P=0.2688 and P=0.1304). CONCLUSION Obese Zucker rats display intracranial hypertension and increased AQP1 expression in CP compared to lean controls. The mechanisms behind these changes are still unknown, but appear to be unrelated to altered pCO2 levels or retinol metabolism. This indicates that the increase in ICP might be related to increased AQP1 levels in CP. Although further studies are warranted, obese Zucker rats could potentially model some aspects of the IIH pathophysiology.
Collapse
Affiliation(s)
- M Uldall
- Department of Neurology, Danish Headache Center, Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark.,Glostrup Research Institute, Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark
| | - D K Bhatt
- Department of Neurology, Danish Headache Center, Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark.,Glostrup Research Institute, Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark
| | - C Kruuse
- Department of Neurology, Neurovascular Research Unit, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - M Juhler
- Department of Neurosurgery, Rigshospitalet Blegdamsvej, University of Copenhagen, Copenhagen, Denmark
| | - I Jansen-Olesen
- Department of Neurology, Danish Headache Center, Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark.,Glostrup Research Institute, Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark
| | - R H Jensen
- Department of Neurology, Danish Headache Center, Rigshospitalet Glostrup, University of Copenhagen, Glostrup, Denmark
| |
Collapse
|
103
|
Tups A, Benzler J, Sergi D, Ladyman SR, Williams LM. Central Regulation of Glucose Homeostasis. Compr Physiol 2017; 7:741-764. [PMID: 28333388 DOI: 10.1002/cphy.c160015] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
104
|
Heinrich G, Muturi HT, Rezaei K, Al-Share QY, DeAngelis AM, Bowman TA, Ghadieh HE, Ghanem SS, Zhang D, Garofalo RS, Yin L, Najjar SM. Reduced Hepatic Carcinoembryonic Antigen-Related Cell Adhesion Molecule 1 Level in Obesity. Front Endocrinol (Lausanne) 2017; 8:54. [PMID: 28396653 PMCID: PMC5366977 DOI: 10.3389/fendo.2017.00054] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 03/03/2017] [Indexed: 12/12/2022] Open
Abstract
Impairment of insulin clearance is being increasingly recognized as a critical step in the development of insulin resistance and metabolic disease. The carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) promotes insulin clearance. Null deletion or liver-specific inactivation of Ceacam1 in mice causes a defect in insulin clearance, insulin resistance, steatohepatitis, and visceral obesity. Immunohistological analysis revealed reduction of hepatic CEACAM1 in obese subjects with fatty liver disease. Thus, we aimed to determine whether this occurs at the hepatocyte level in response to systemic extrahepatic factors and whether this holds across species. Northern and Western blot analyses demonstrate that CEACAM1 mRNA and protein levels are reduced in liver tissues of obese individuals compared to their lean age-matched counterparts. Furthermore, Western analysis reveals a comparable reduction of CEACAM1 protein in primary hepatocytes derived from the same obese subjects. Similar to humans, Ceacam1 mRNA level, assessed by quantitative RT-PCR analysis, is significantly reduced in the livers of obese Zucker (fa/fa, ZDF) and Koletsky (f/f) rats relative to their age-matched lean counterparts. These studies demonstrate that the reduction of hepatic CEACAM1 in obesity occurs at the level of hepatocytes and identify the reduction of hepatic CEACAM1 as a common denominator of obesity across multiple species.
Collapse
Affiliation(s)
- Garrett Heinrich
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Harrison T. Muturi
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Khadijeh Rezaei
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Qusai Y. Al-Share
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Anthony M. DeAngelis
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Thomas A. Bowman
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Hilda E. Ghadieh
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Simona S. Ghanem
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
| | - Deqiang Zhang
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Lei Yin
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sonia M. Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Center for Diabetes and Endocrine Research (CeDER), College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA
- *Correspondence: Sonia M. Najjar,
| |
Collapse
|
105
|
Cardoso AM, Alves MG, Mathur PP, Oliveira PF, Cavaco JE, Rato L. Obesogens and male fertility. Obes Rev 2017; 18:109-125. [PMID: 27776203 DOI: 10.1111/obr.12469] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/04/2016] [Accepted: 08/15/2016] [Indexed: 12/28/2022]
Abstract
In the last decades, several studies evidenced a decrease in male fertility in developed countries. Although the aetiology of this trend in male reproductive health remains a matter of debate, environmental compounds that predispose to weight gain, namely obesogens, are appointed as contributors because of their action as endocrine disruptors. Obesogens favour adipogenesis by an imbalance of metabolic processes and can be found virtually everywhere. These compounds easily accumulate in tissues with high lipid content. Obesogens change the functioning of male reproductive axis, and, consequently, the testicular physiology and metabolism that are pivotal for spermatogenesis. The disruption of these tightly regulated metabolic pathways leads to adverse reproductive outcomes. Notably, adverse effects of obesogens may also promote disturbances in the metabolic performance of the following generations, through epigenetic modifications passed by male gametes. Thus, unveiling the molecular pathways by which obesogens induce toxicity that may end up in epigenetic modifications is imperative. Otherwise, a transgenerational susceptibility to metabolic diseases may be favoured. We present an up-to-date overview of the impact of obesogens on testicular physiology, with a particular focus on testicular metabolism. We also address the effects of obesogens on male reproductive parameters and the subsequent consequences for male fertility.
Collapse
Affiliation(s)
- A M Cardoso
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| | - M G Alves
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| | - P P Mathur
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, India.,KIIT University, Bhubaneswar, India
| | - P F Oliveira
- Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS) and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Porto, Portugal.,i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - J E Cavaco
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| | - L Rato
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
106
|
Burnett LC, Skowronski AA, Rausch R, LeDuc CA, Leibel RL. Determination of the half-life of circulating leptin in the mouse. Int J Obes (Lond) 2016; 41:355-359. [PMID: 28025576 PMCID: PMC5340585 DOI: 10.1038/ijo.2016.238] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/16/2016] [Accepted: 12/11/2016] [Indexed: 01/01/2023]
Abstract
Background The adipokine hormone, leptin, is a major component of body weight homeostasis. Numerous studies have been performed administering recombinant mouse leptin as an experimental reagent; however, the half life of circulating leptin following exogenous administration of recombinant mouse leptin has not been carefully evaluated. Methods Exogenous leptin was administered (3 mg leptin/kg body weight) to ten week old fasted non-obese male mice and plasma was serially collected at seven time points; plasma leptin concentration was measured by ELISA at each time point to estimate the circulating half life of mouse leptin. Results Under the physiological circumstances tested, the half life of mouse leptin was 40.2 (+/− 2.2) minutes. Circulating leptin concentrations up to one hour following exogenous leptin administration were 170-fold higher than endogenous levels at fasting. Conclusions The half life of mouse leptin was determined to be 40.2 minutes. These results should be useful in planning and interpreting experiments employing exogenous leptin. The unphysiological elevations in circulating leptin resulting from widely used dosing regimens for exogenous leptin are likely to confound inferences regarding some aspects of the hormone’s clinical biology.
Collapse
Affiliation(s)
- L C Burnett
- Columbia University Institute of Human Nutrition, New York, NY, USA.,Columbia University Department of Pediatrics, Division of Molecular Genetics, New York, NY, USA.,Naomi Berrie Diabetes Center, New York, NY, USA
| | - A A Skowronski
- Columbia University Institute of Human Nutrition, New York, NY, USA.,Columbia University Department of Pediatrics, Division of Molecular Genetics, New York, NY, USA.,Naomi Berrie Diabetes Center, New York, NY, USA
| | - R Rausch
- Columbia University Department of Pediatrics, Division of Molecular Genetics, New York, NY, USA.,Naomi Berrie Diabetes Center, New York, NY, USA
| | - C A LeDuc
- Columbia University Department of Pediatrics, Division of Molecular Genetics, New York, NY, USA.,Naomi Berrie Diabetes Center, New York, NY, USA.,New York Obesity Research Center, New York, NY, USA
| | - R L Leibel
- Columbia University Department of Pediatrics, Division of Molecular Genetics, New York, NY, USA.,Naomi Berrie Diabetes Center, New York, NY, USA.,New York Obesity Research Center, New York, NY, USA
| |
Collapse
|
107
|
Wasik AA, Dumont V, Tienari J, Nyman TA, Fogarty CL, Forsblom C, Lehto M, Lehtonen E, Groop PH, Lehtonen S. Septin 7 reduces nonmuscle myosin IIA activity in the SNAP23 complex and hinders GLUT4 storage vesicle docking and fusion. Exp Cell Res 2016; 350:336-348. [PMID: 28011197 PMCID: PMC5243148 DOI: 10.1016/j.yexcr.2016.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 12/10/2016] [Accepted: 12/17/2016] [Indexed: 12/28/2022]
Abstract
Glomerular epithelial cells, podocytes, are insulin responsive and can develop insulin resistance. Here, we demonstrate that the small GTPase septin 7 forms a complex with nonmuscle myosin heavy chain IIA (NMHC-IIA; encoded by MYH9), a component of the nonmuscle myosin IIA (NM-IIA) hexameric complex. We observed that knockdown of NMHC-IIA decreases insulin-stimulated glucose uptake into podocytes. Both septin 7 and NM-IIA associate with SNAP23, a SNARE protein involved in GLUT4 storage vesicle (GSV) docking and fusion with the plasma membrane. We observed that insulin decreases the level of septin 7 and increases the activity of NM-IIA in the SNAP23 complex, as visualized by increased phosphorylation of myosin regulatory light chain. Also knockdown of septin 7 increases the activity of NM-IIA in the complex. The activity of NM-IIA is increased in diabetic rat glomeruli and cultured human podocytes exposed to macroalbuminuric sera from patients with type 1 diabetes. Collectively, the data suggest that the activity of NM-IIA in the SNAP23 complex plays a key role in insulin-stimulated glucose uptake into podocytes. Furthermore, we observed that septin 7 reduces the activity of NM-IIA in the SNAP23 complex and thereby hinders GSV docking and fusion with the plasma membrane. Septin 7, nonmuscle myosin heavy chain IIA (NMHC-IIA) and SNAP23 form a complex. Knockdown of septin 7 increases NM-IIA activity in the SNAP23 complex. Insulin decreases septin 7 level and increases NM-IIA activity in the SNAP23 complex. Septin 7 hinders GSV docking/fusion by reducing NM-IIA activity in the SNAP23 complex.
Collapse
Affiliation(s)
- Anita A Wasik
- Department of Pathology, University of Helsinki, 00014 Helsinki, Finland
| | - Vincent Dumont
- Department of Pathology, University of Helsinki, 00014 Helsinki, Finland
| | - Jukka Tienari
- Department of Pathology, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, 05850 Hyvinkää, Finland
| | - Tuula A Nyman
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| | - Christopher L Fogarty
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, 00290 Helsinki, Finland; Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, 000290 Helsinki, Finland; Diabetes&Obesity Research Program, Research Program´s Unit, 00014 University of Helsinki, Finland
| | - Carol Forsblom
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, 00290 Helsinki, Finland; Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, 000290 Helsinki, Finland; Diabetes&Obesity Research Program, Research Program´s Unit, 00014 University of Helsinki, Finland
| | - Markku Lehto
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, 00290 Helsinki, Finland; Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, 000290 Helsinki, Finland; Diabetes&Obesity Research Program, Research Program´s Unit, 00014 University of Helsinki, Finland
| | - Eero Lehtonen
- Department of Pathology, University of Helsinki, 00014 Helsinki, Finland; Laboratory Animal Centre, University of Helsinki, 00014 Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, 00290 Helsinki, Finland; Abdominal Center Nephrology, University of Helsinki and Helsinki University Hospital, 000290 Helsinki, Finland; Diabetes&Obesity Research Program, Research Program´s Unit, 00014 University of Helsinki, Finland; Baker IDI Heart & Diabetes Institute, 3004 Melbourne, Australia
| | - Sanna Lehtonen
- Department of Pathology, University of Helsinki, 00014 Helsinki, Finland.
| |
Collapse
|
108
|
Comparison of cocaine reinforcement in lean and obese Zucker rats: Relative potency and reinstatement of extinguished operant responding. Physiol Behav 2016; 170:88-92. [PMID: 27998754 DOI: 10.1016/j.physbeh.2016.12.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/14/2016] [Accepted: 12/13/2016] [Indexed: 11/23/2022]
Abstract
AIMS Evidence indicates that obese individuals exhibit alterations in brain-reward function that are anatomically and functionally similar to what has been observed in drug addicts, which could theoretically make obese individuals vulnerable to drug abuse and drug abusers vulnerable to overeating. However, few studies have investigated the cross-generality of these phenotypes. We recently reported that the reinforcing effectiveness (i.e., value) of a fat was greater in obese Zucker rats than in their lean counterparts, but found no differences in the reinforcing effectiveness of cocaine between groups, suggesting psychostimulant reinforcement is similar in lean and obese Zucker rats. However, it is unknown if other aspects of reinforcement such as cocaine's potency as a reinforcer or its reinstating effects differ in lean and obese Zucker rats. METHODS The current study compared cocaine's potency as a reinforcer in lean and obese Zucker rats self-administering intravenous cocaine (0.06-1.0mg/kg/inj), and subsequently tested these subjects in cue- (light) and drug-primed (intraperitoneal cocaine; 10mg/kg) reinstatement of extinguished operant responding. RESULTS All rats acquired cocaine self-administration and generated "inverted-U" dose-response functions. Following extinction of responding, the cue- and drug-primes increased lever-pressing in both groups (i.e., reinstatement). No significant differences in the reinforcing potency or reinstating effects of cocaine were observed as a function of obesity. CONCLUSIONS These results, combined with our previous observations, demonstrate that cocaine's reinforcing effects are comparable in lean and obese Zucker rats and do not support the hypothesis that obesity is associated with an altered reinforcing effect of psychostimulants.
Collapse
|
109
|
Fasting selectively blocks development of acute lymphoblastic leukemia via leptin-receptor upregulation. Nat Med 2016; 23:79-90. [PMID: 27941793 DOI: 10.1038/nm.4252] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/14/2016] [Indexed: 12/17/2022]
Abstract
New therapeutic approaches are needed to treat leukemia effectively. Dietary restriction regimens, including fasting, have been considered for the prevention and treatment of certain solid tumor types. However, whether and how dietary restriction affects hematopoietic malignancies is unknown. Here we report that fasting alone robustly inhibits the initiation and reverses the leukemic progression of both B cell and T cell acute lymphoblastic leukemia (B-ALL and T-ALL, respectively), but not acute myeloid leukemia (AML), in mouse models of these tumors. Mechanistically, we found that attenuated leptin-receptor (LEPR) expression is essential for the development and maintenance of ALL, and that fasting inhibits ALL development by upregulation of LEPR and its downstream signaling through the protein PR/SET domain 1 (PRDM1). The expression of LEPR signaling-related genes correlated with the prognosis of pediatric patients with pre-B-ALL, and fasting effectively inhibited B-ALL growth in a human xenograft model. Our results indicate that the effects of fasting on tumor growth are cancer-type dependent, and they suggest new avenues for the development of treatment strategies for leukemia.
Collapse
|
110
|
Cleary MP, Juneja SC, Phillips FC, Hu X, Grande JP, Maihle NJ. Leptin Receptor-Deficient MMTV-TGF-α/Leprdb Leprdb Female Mice Do Not Develop Oncogene-Induced Mammary Tumors. Exp Biol Med (Maywood) 2016; 229:182-93. [PMID: 14734797 DOI: 10.1177/153537020422900207] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Being overweight is a risk factor for postmenopausal breast cancer and is associated with an increased incidence and shortened latency of spontaneous and chemically Induced mammary tumors in rodents. However, leptin-deficient obese Lepob Lepob female mice have reduced incidences of spontaneous and oncogene-induced mammary tumors. Of interest, leptin enhances the proliferation of human breast cancer cell lines in which leptin receptors are expressed, which suggests that leptin signaling plays a role in tumor development. We evaluated oncogene-induced mammary tumor development in obese MMTV-TGF-α/Leprdb Leprdb mice that exhibit a defect in OB-Rb, which is considered to be the major signaling isoform of the leptin receptor. Lepr and MMTV-TGF-α mice were crossed, and the offspring were genotyped for oncogene expression and the determination of Lepr status. Lean MMTV-TGF-α/Lepr+ Lepr+ (homozygous) and MMTV-TGF-α/Lepr+ Leprdb (heterozygous) mice and obese MMTV-TGF-α/Leprdb Leprdb mice were monitored until age 104 weeks. Body weights of MMTV-TGF-α/Leprdb Leprdb mice were significantly heavier than those of the lean groups. No mammary tumors were detected in MMTV-TGF-α/LeprdbLeprdb mice, whereas the incidence of mammary tumors in MMTV-TGF-α/Lepr+ Lepr+ and MMTV-TGF-α/Lepr+ Leprdb mice was 69% and 82%, respectively. Examination of mammary tissue whole mounts indicated an absence of duct formation and branching for MMTV-TGF-α/Leprdb Leprdb mice. Both age at mammary tumor detection and tumor burden (tumors/mouse and tumor weights) were similar for the lean genotypes. Serum leptin levels of MMTV-TGF-α/Leprdb Leprdb mice were 12-20-fold higher than levels of lean mice. Thus, despite elevated serum leptin levels, leptin receptor-deficient MMTV-TGF-α/Leprdb Leprdb mice do not develop mammary tumors. This study provides additional evidence that leptin and its cognate receptor may be involved in mammary tumorigenesis.
Collapse
MESH Headings
- Animals
- Body Weight
- Disease Models, Animal
- Female
- Leptin/blood
- Leptin/metabolism
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Obesity/genetics
- Obesity/pathology
- Oncogenes/genetics
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/physiology
- Receptors, Leptin
- Survival Analysis
- Transforming Growth Factor alpha/genetics
Collapse
Affiliation(s)
- Margot P Cleary
- Hormel Institute, University of Minnesota, Austin, Minnesota 55912, USA.
| | | | | | | | | | | |
Collapse
|
111
|
Abstract
Multiple physiologic and neural systems contribute to the controls over what and how much we eat. These systems include signaling involved in the detection and signaling of nutrient availability, signals arising from consumed nutrients that provide feedback information during a meal to induce satiation, and signals related to the rewarding properties of eating. Each of these has a separate neural representation, but important interactions among these systems are critical to the overall controls of food intake.
Collapse
Affiliation(s)
- Timothy H Moran
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA; Global Obesity Prevention Center at Johns Hopkins, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA.
| | - Ellen E Ladenheim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|
112
|
Sakata T, Yoshimatsu H, Masaki T, Tsuda K. Anti-Obesity Actions of Mastication Driven by Histamine Neurons in Rats. Exp Biol Med (Maywood) 2016; 228:1106-10. [PMID: 14610247 DOI: 10.1177/153537020322801002] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Implications of mastication in energy intake and expenditure regulated by histamine (HA) neurons were investigated in rats. Depletion of neuronal HA from the mesencephalic trigeminal sensory nucleus (Me5) reduced eating speed, but that from a satiety center of the ventromedial hypothalamus (VMH) increased both meal size and its duration leaving eating speed unaffected. Turnover of neuronal HA in the Me5 was elevated at the early phase of feeding and that in the VMH was at the later phase. This elevated turnover was abolished by gastric intubations of an isocaloric liquid diet or an equivolume of water. Mastication-induced activation of HA neurons suppressed physiological food intake through H1-receptor in the hypothalamic paraventricular nucleus (PVN) and the VMH. On the other hand, the HA neurons activation accelerated lipolysis particularly in the visceral adipose tissues and up-regulated mRNA expression of uncoupling protein family through sympathetic efferent nerve. Mastication thus plays an important role as a potent input signal to activate HA neurons. Our recent findings have evidently shown how tightly and elegantly HA neurons are concordant with leptin signaling system through a negative feedback loop.
Collapse
Affiliation(s)
- Toshiie Sakata
- Department of Nutritional Sciences, Faculty of Nutritional Sciences, Nakamura Gakuen University, Fukuoka, 814-0198 Japan.
| | | | | | | |
Collapse
|
113
|
Beck B, Richy S, Stricker-Krongrad A. Ghrelin and Body Weight Regulation in the Obese Zucker Rat in Relation to Feeding State and Dark/Light Cycle. Exp Biol Med (Maywood) 2016; 228:1124-31. [PMID: 14610250 DOI: 10.1177/153537020322801005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Ghrelin is a new orexigenic peptide primarily produced by the stomach but also present in the hypothalamus. It has adipogenic effects when it is chronically injected in rodents but in obese humans, its plasma concentration is decreased. It can reverse the anorectic effects of leptin when it is co-injected with this peptide in the brain ventricles. The Zucker fa/fa rat is a genetic model of obesity related to a default in the leptin receptor. It is characterized by a large dysregulation of numerous hypothalamic peptides but the ghrelin status of this rat has not yet been determined. Through several experiments, we determine in lean and obese Zucker rats its circulating form in the plasma, its tissue levels and/or expression, and studied the influence of different feeding conditions and its light/dark variations. Ghrelin expression was higher in the obese stomach and hypothalamus (P < 0.05 and P < 0.02, respectively). The ratio of [Octanoyl-Ser3]-ghrelin (active form) to [Des-Octanoyl-Ser3]-ghrelin (inactive form) was approximately 1:1 in the stomach and 2:1 in the plasma in lean and obese rats (no differences). After fasting, plasma ghrelin concentrations increased significantly in lean (+ 64%; P < 0.001) and obese (+ 60%; P < 0.02) rats. After 24 hours of refeeding, they returned to their initial ad lib levels. Ghrelin concentrations were higher in obese rats by 69% (P < 0.005), 65% (P < 0.02), and 73% (P < 0.005) in the ad libitum, fast, and refed states respectively. These results indicate that the obese Zucker rat is characterized by increases in the stomach mRNA expression and in peptide release in the circulation. They clearly support a role for ghrelin in the development of obesity in the absence of leptin signaling.
Collapse
Affiliation(s)
- Bernard Beck
- Systèmes Neuromodulateurs des Comportements Ingestifs, Nancy, France.
| | | | | |
Collapse
|
114
|
Ferrario CR, Labouèbe G, Liu S, Nieh EH, Routh VH, Xu S, O'Connor EC. Homeostasis Meets Motivation in the Battle to Control Food Intake. J Neurosci 2016; 36:11469-11481. [PMID: 27911750 PMCID: PMC5125214 DOI: 10.1523/jneurosci.2338-16.2016] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 08/30/2016] [Accepted: 09/05/2016] [Indexed: 01/09/2023] Open
Abstract
Signals of energy homeostasis interact closely with neural circuits of motivation to control food intake. An emerging hypothesis is that the transition to maladaptive feeding behavior seen in eating disorders or obesity may arise from dysregulation of these interactions. Focusing on key brain regions involved in the control of food intake (ventral tegmental area, striatum, hypothalamus, and thalamus), we describe how activity of specific cell types embedded within these regions can influence distinct components of motivated feeding behavior. We review how signals of energy homeostasis interact with these regions to influence motivated behavioral output and present evidence that experience-dependent neural adaptations in key feeding circuits may represent cellular correlates of impaired food intake control. Future research into mechanisms that restore the balance of control between signals of homeostasis and motivated feeding behavior may inspire new treatment options for eating disorders and obesity.
Collapse
Affiliation(s)
- Carrie R Ferrario
- University of Michigan Medical School, Department of Pharmacology, Ann Arbor, Michigan 48109-5632
| | - Gwenaël Labouèbe
- University of Lausanne, Center for Integrative Genomics, Lausanne, CH1015, Switzerland
| | - Shuai Liu
- University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Edward H Nieh
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | | | - Shengjin Xu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia 20147, and
| | - Eoin C O'Connor
- University of Geneva, Department of Basic Neuroscience, Geneva, CH1211, Switzerland
| |
Collapse
|
115
|
Srivastava A, Srivastava N, Mittal B. Genetics of Obesity. Indian J Clin Biochem 2016; 31:361-371. [PMID: 27605733 PMCID: PMC4992482 DOI: 10.1007/s12291-015-0541-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 12/08/2015] [Indexed: 12/29/2022]
Abstract
Numerous classical genetic studies have proved that genes are contributory factors for obesity. Genes are directly responsible for obesity associated disorders such as Bardet-Biedl and Prader-Willi syndromes. However, both genes as well as environment are associated with obesity in the general population. Genetic epidemiological approaches, particularly genome-wide association studies, have unraveled many genes which play important roles in human obesity. Elucidation of their biological functions can be very useful for understanding pathobiology of obesity. In the near future, further exploration of obesity genetics may help to develop useful diagnostic and predictive tests for obesity treatment.
Collapse
Affiliation(s)
- Apurva Srivastava
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Bareli Road, Lucknow, Uttar Pradesh 226014 India
- Department of Physiology, King George’s Medical University, Chowk, Lucknow, Uttar Pradesh 226003 India
| | - Neena Srivastava
- Department of Physiology, King George’s Medical University, Chowk, Lucknow, Uttar Pradesh 226003 India
| | - Balraj Mittal
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Rae Bareli Road, Lucknow, Uttar Pradesh 226014 India
| |
Collapse
|
116
|
Functional Differences of Very-Low-Density Lipoprotein Receptor Splice Variants in Regulating Wnt Signaling. Mol Cell Biol 2016; 36:2645-54. [PMID: 27528615 PMCID: PMC5038150 DOI: 10.1128/mcb.00235-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/22/2016] [Indexed: 11/20/2022] Open
Abstract
The very-low-density lipoprotein receptor (VLDLR) negatively regulates Wnt signaling. VLDLR has two major alternative splice variants, VLDLRI and VLDLRII, but their biological significance and distinction are unknown. Here we found that most tissues expressed both VLDLRI and VLDLRII, while the retina expressed only VLDLRII. The shed soluble VLDLR extracellular domain (sVLDLR-N) was detected in the conditioned medium of retinal pigment epithelial cells, interphotoreceptor matrix, and mouse plasma, indicating that ectodomain shedding of VLDLR occurs endogenously. VLDLRII displayed a higher ectodomain shedding rate and a more potent inhibitory effect on Wnt signaling than VLDLRI in vitro and in vivo O-glycosylation, which is present in VLDLRI but not VLDLRII, determined the differential ectodomain shedding rates. Moreover, the release of sVLDLR-N was inhibited by a metalloproteinase inhibitor, TAPI-1, while it was promoted by phorbol 12-myristate 13-acetate (PMA). In addition, sVLDLR-N shedding was suppressed under hypoxia. Further, plasma levels of sVLDLR-N were reduced in both type 1 and type 2 diabetic mouse models. We concluded that VLDLRI and VLDLRII had differential roles in regulating Wnt signaling and that decreased plasma levels of sVLDLR-N may contribute to Wnt signaling activation in diabetic complications. Our study reveals a novel mechanism for intercellular regulation of Wnt signaling through VLDLR ectodomain shedding.
Collapse
|
117
|
Kawano Y, Nakae J, Watanabe N, Kikuchi T, Tateya S, Tamori Y, Kaneko M, Abe T, Onodera M, Itoh H. Colonic Pro-inflammatory Macrophages Cause Insulin Resistance in an Intestinal Ccl2/Ccr2-Dependent Manner. Cell Metab 2016; 24:295-310. [PMID: 27508875 DOI: 10.1016/j.cmet.2016.07.009] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 03/22/2016] [Accepted: 07/15/2016] [Indexed: 12/27/2022]
Abstract
High-fat diet (HFD) induces low-grade chronic inflammation and insulin resistance. However, little is known about the mechanism underlying HFD-induced chronic inflammation in peripheral insulin-responsive tissues. Here, we show that colonic pro-inflammatory macrophages regulate insulin sensitivity under HFD conditions. To investigate the pathophysiological role of colonic macrophages, we generated macrophage-specific chemokine (C-C Motif) receptor 2 (Ccr2) knockout (M-Ccr2KO) and intestinal epithelial cell-specific tamoxifen-inducible Ccl2 knockout (Vil-Ccl2KO) mice. Both strains exhibited similar body weight to control under HFD. However, they exhibited decreased infiltration of colonic pro-inflammatory macrophages, decreased intestinal permeability, and inactivation of the colonic inflammasome. Interestingly, they showed significantly improved glucose tolerance and insulin sensitivity with decreased chronic inflammation of adipose tissue. Therefore, inhibition of pro-inflammatory macrophage infiltration prevents HFD-induced insulin resistance and could be a novel therapeutic approach for type 2 diabetes.
Collapse
Affiliation(s)
- Yoshinaga Kawano
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Jun Nakae
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan.
| | - Nobuyuki Watanabe
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-0074, Japan
| | - Tetsuhiro Kikuchi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Sanshiro Tateya
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yoshikazu Tamori
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Department of Endocrinology and Metabolism, Chibune General Hospital, Osaka 555-0011, Japan
| | - Mari Kaneko
- Animal Resource Development Unit, Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies, Kobe 650-0047, Japan; Genetic Engineering Team, Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies, Kobe 650-0047, Japan
| | - Takaya Abe
- Genetic Engineering Team, Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies, Kobe 650-0047, Japan
| | - Masafumi Onodera
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-0074, Japan
| | - Hiroshi Itoh
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
118
|
Astragaloside IV improves lipid metabolism in obese mice by alleviation of leptin resistance and regulation of thermogenic network. Sci Rep 2016; 6:30190. [PMID: 27444146 PMCID: PMC4957129 DOI: 10.1038/srep30190] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/30/2016] [Indexed: 02/07/2023] Open
Abstract
Obesity is a worldwide threat to public health in modern society, which may result from leptin resistance and disorder of thermogenesis. The present study investigated whether astragaloside IV (ASI) could prevent obesity in high-fat diet (HFD)-fed and db/db mice. In HFD-fed mice, ASI prevented body weight gain, lowered serum triglyceride and total cholesterol levels, mitigated liver lipid accumulation, reduced fat tissues and decreased the enlargement of adipose cells. In metabolic chambers, ASI lessened appetite of the mice, decreased their respiratory exchange ratio and elevated VCO2 and VO2 without altering circadian motor activity. Moreover, ASI modulated thermogenesis associated gene expressions in liver and brawn fat tissues, as well as leptin resistance evidenced by altered expressions of leptin, leptin receptor (ObR) or appetite associated genes. In SH-SY5Y cells, ASI enhanced leptin signaling transduction. However, in db/db mice, ASI did not change body weight gain and appetite associated genes. But it decreased serum triglyceride and total cholesterol levels as well as liver triglyceride. Meanwhile, it significantly modulated gene expressions of PPARα, PGC1-α, UCP2, ACC, SCD1, LPL, AP2, CD36 and SREBP-1c. Collectively, our study suggested that ASI could efficiently improve lipid metabolism in obese mice probably through enhancing leptin sensitivity and modulating thermogenic network.
Collapse
|
119
|
Uehara Y, Shimizu H, Ohtani KI, Tsuchiya T, Takahashi H, Sato N, Shimomura Y, Mori M. Relationship between Hypothalamic—Pituitary—Adrenal Axis Function and Leptin Release. Nutr Neurosci 2016; 1:77-82. [DOI: 10.1080/1028415x.1998.11747215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
120
|
Alliouachene S, Bilanges B, Chaussade C, Pearce W, Foukas LC, Scudamore CL, Moniz LS, Vanhaesebroeck B. Inactivation of class II PI3K-C2α induces leptin resistance, age-dependent insulin resistance and obesity in male mice. Diabetologia 2016; 59:1503-1512. [PMID: 27138914 PMCID: PMC4901096 DOI: 10.1007/s00125-016-3963-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 03/24/2016] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS While the class I phosphoinositide 3-kinases (PI3Ks) are well-documented positive regulators of metabolism, the involvement of class II PI3K isoforms (PI3K-C2α, -C2β and -C2γ) in metabolic regulation is just emerging. Organismal inactivation of PI3K-C2β increases insulin signalling and sensitivity, whereas PI3K-C2γ inactivation has a negative metabolic impact. In contrast, the role of PI3K-C2α in organismal metabolism remains unexplored. In this study, we investigated whether kinase inactivation of PI3K-C2α affects glucose metabolism in mice. METHODS We have generated and characterised a mouse line with a constitutive inactivating knock-in (KI) mutation in the kinase domain of the gene encoding PI3K-C2α (Pik3c2a). RESULTS While homozygosity for kinase-dead PI3K-C2α was embryonic lethal, heterozygous PI3K-C2α KI mice were viable and fertile, with no significant histopathological findings. However, male heterozygous mice showed early onset leptin resistance, with a defect in leptin signalling in the hypothalamus, correlating with a mild, age-dependent obesity, insulin resistance and glucose intolerance. Insulin signalling was unaffected in insulin target tissues of PI3K-C2α KI mice, in contrast to previous reports in which downregulation of PI3K-C2α in cell lines was shown to dampen insulin signalling. Interestingly, no metabolic phenotypes were detected in female PI3K-C2α KI mice at any age. CONCLUSIONS/INTERPRETATION Our data uncover a sex-dependent role for PI3K-C2α in the modulation of hypothalamic leptin action and systemic glucose homeostasis. ACCESS TO RESEARCH MATERIALS All reagents are available upon request.
Collapse
Affiliation(s)
- Samira Alliouachene
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
| | - Benoit Bilanges
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
| | - Claire Chaussade
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
- Galderma R&D, Sophia Antipolis Cedex, France
| | - Wayne Pearce
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Lazaros C Foukas
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Cheryl L Scudamore
- Mary Lyon Centre, MRC Harwell, Harwell Science and Innovation Campus, Harwell, UK
| | - Larissa S Moniz
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Bart Vanhaesebroeck
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
| |
Collapse
|
121
|
Ishihara A, Matsumoto E, Horikawa K, Kudo T, Sakao E, Nemoto A, Iwase K, Sugiyama H, Tamura Y, Shibata S, Takiguchi M. Multifactorial Regulation of Daily Rhythms in Expression of the Metabolically Responsive Gene Spot14 in the Mouse Liver. J Biol Rhythms 2016; 22:324-34. [PMID: 17660449 DOI: 10.1177/0748730407302107] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Spot14 is a putative transcriptional regulator for genes involved in fatty acid synthesis. The Spot14 gene is activated in response to lipogenic stimuli such as dietary carbohydrate and is also under circadian regulation. The authors investigated factors responsible for daily oscillation of Spot14 expression. If mice were kept under a 12-h light/12-h dark cycle with ad libitum feeding, Spot14 mRNA levels in the liver reached a peak at an early dark period when mice, as nocturnal animals, start feeding. Under fasting, while Spot14 mRNA levels were generally decreased, the rhythmicity was still maintained, suggesting contribution of both nutritional elements and circadian clock factors on robust rhythmicity of Spot14 expression. Effects of circadian clock factors were confirmed by the observations that the circadian rhythm of Spot14 expression was seen also under the constant darkness and that the rhythmicity was lost in Clock mutant mice. When mice were housed in short-photoperiod (6-h light/18-h dark) and long-photoperiod (18-h light/6-h dark) cycles, rhythms of Spot14 mRNA levels were phase advanced and phase delayed, respectively, being concordant with the notion that Spot14 expression is under the control of the light-entrainable oscillator. As for nutritional mediators, in the liver of db/ db mice exhibiting hyperinsulinemia-accompanied hyperglycemia, Spot14 mRNA levels were constantly high without apparent rhythmicity, consistent with previous observations for strong activation of the Spot14 gene by glucose and insulin. Restricted feeding during the 4-h mid-light period caused a phase advance of the Spot14 expression rhythm. On the other hand, restricted feeding during the 4-h mid-dark period led to damping of the rhythmicity, apparently resulting from the separation of phases between effects of the light/dark cycle and feeding on Spot14 expression. Thus, the daily rhythm of Spot14 expression in the liver is under the control of the light-entrainable oscillator, food-entrainable oscillator, and food-derived nutrients, in a separate or cooperative manner.
Collapse
Affiliation(s)
- Akinori Ishihara
- Department of Biochemistry and Genetics, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Mela V, Vargas A, Meza C, Kachani M, Wagner EJ. Modulatory influences of estradiol and other anorexigenic hormones on metabotropic, Gi/o-coupled receptor function in the hypothalamic control of energy homeostasis. J Steroid Biochem Mol Biol 2016; 160:15-26. [PMID: 26232394 PMCID: PMC4732935 DOI: 10.1016/j.jsbmb.2015.07.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/22/2015] [Accepted: 07/26/2015] [Indexed: 11/22/2022]
Abstract
The appetite suppressant actions of estradiol are due to its ability to attenuate orexigenic signals and potentiate anorexigenic signals. The work from my laboratory has shown that male guinea pigs are more sensitive to the hyperphagic and hypothermic effects of cannabinoids than their female counterparts. Cannabinoid sensitivity is further dampened by the activational effects of estradiol. This occurs via the hypothalamic feeding circuitry, where estradiol rapidly attenuates the cannabinoid CB1 receptor-mediated presynaptic inhibition of glutamatergic input onto anorexigenic proopiomelanocortin (POMC) neurons in the arcuate nucleus. This disruption is blocked by the estrogen receptor antagonist ICI 182,780, and associated with increased expression of phosphatidylinositol-3-kinase (PI3K). Moreover, the ability of estradiol to reduce both the cannabinoid-induced hyperphagia and glutamate release onto POMC neurons is abrogated by the PI3K inhibitor PI 828. The peptide orphanin FQ/nociceptin (OFQ/N) activates opioid receptor-like (ORL)1 receptors to hyperpolarize and inhibit POMC neurons via the activation of postsynaptic G protein-gated, inwardly-rectifying (GIRK) channels. We have demonstrated that the fasting-induced hyperphagia observed in ORL1-null mice is blunted compared to wild type controls. In addition, the ORL1 receptor-mediated activation of GIRK channels in POMC neurons from ovariectomized female rats is markedly impaired by estradiol. The estrogenic attenuation of presynaptic CB1 and postsynaptic ORL1 receptor function may be part of a more generalized mechanism through which anorexigenic hormones suppress orexigenic signaling. Indeed, we have found that leptin robustly suppresses the OFQ/N-induced activation of GIRK channels in POMC neurons. Furthermore, its ability to augment excitatory input onto POMC neurons is blocked by PI 828. Thus, estradiol and other hormones like leptin reduce energy intake at least partly by activating PI3K to disrupt the pleiotropic functions of Gi/o-coupled receptors that inhibit anorexigenic POMC neurons.
Collapse
Affiliation(s)
- Virginia Mela
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States
| | - Amanda Vargas
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States
| | - Cecilia Meza
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States
| | - Malika Kachani
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766, United States
| | - Edward J Wagner
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States.
| |
Collapse
|
123
|
Abstract
Obesity and its major comorbidities, including type 2 diabetes mellitus, nonalcoholic fatty liver disease (NAFLD), obesity cardiomyopathy, and certain cancers, have caused life expectancy in the United States to decline in recent years. Obesity is the increased accumulation of triglycerides (TG), which are synthesized from glycerol and long-chain fatty acids (LCFA) throughout the body. LCFA enter adipocytes, hepatocytes, and cardiomyocytes via specific, facilitated transport processes. Metabolism of increased cellular TG content in obesity may lead to comorbidities such as NAFLD and cardiomyopathy. Better understanding of LCFA transport processes may lead to successful treatment of obesity and NAFLD.
Collapse
Affiliation(s)
- Paul D Berk
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia College of Physicians and Surgeons, Columbia University Medical Center, 650 West 168 Street, New York, NY 10032, USA; Division of Preventive Medicine, Department of Medicine, Columbia College of Physicians and Surgeons, Columbia University Medical Center, William Black Building, 650 West 168 Street, Room 1006, Box 57A, New York, NY 10032, USA.
| | - Elizabeth C Verna
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia College of Physicians and Surgeons, Columbia University Medical Center, 650 West 168 Street, New York, NY 10032, USA
| |
Collapse
|
124
|
Cyclin-dependent kinase 2 protects podocytes from apoptosis. Sci Rep 2016; 6:21664. [PMID: 26876672 PMCID: PMC4753499 DOI: 10.1038/srep21664] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 01/14/2016] [Indexed: 12/12/2022] Open
Abstract
Loss of podocytes is an early feature of diabetic nephropathy (DN) and predicts its progression. We found that treatment of podocytes with sera from normoalbuminuric type 1 diabetes patients with high lipopolysaccharide (LPS) activity, known to predict progression of DN, downregulated CDK2 (cyclin-dependent kinase 2). LPS-treatment of mice also reduced CDK2 expression. LPS-induced downregulation of CDK2 was prevented in vitro and in vivo by inhibiting the Toll-like receptor (TLR) pathway using immunomodulatory agent GIT27. We also observed that CDK2 is downregulated in the glomeruli of obese Zucker rats before the onset of proteinuria. Knockdown of CDK2, or inhibiting its activity with roscovitine in podocytes increased apoptosis. CDK2 knockdown also reduced expression of PDK1, an activator of the cell survival kinase Akt, and reduced Akt phosphorylation. This suggests that CDK2 regulates the activity of the cell survival pathway via PDK1. Furthermore, PDK1 knockdown reduced the expression of CDK2 suggesting a regulatory loop between CDK2 and PDK1. Collectively, our data show that CDK2 protects podocytes from apoptosis and that reduced expression of CDK2 associates with the development of DN. Preventing downregulation of CDK2 by blocking the TLR pathway with GIT27 may provide a means to prevent podocyte apoptosis and progression of DN.
Collapse
|
125
|
Trasino SE, Tang XH, Jessurun J, Gudas LJ. Retinoic acid receptor β2 agonists restore glycaemic control in diabetes and reduce steatosis. Diabetes Obes Metab 2016; 18:142-51. [PMID: 26462866 PMCID: PMC4948868 DOI: 10.1111/dom.12590] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 09/22/2015] [Accepted: 10/09/2015] [Indexed: 01/09/2023]
Abstract
AIMS To investigate the effects of specific retinoic acid receptor (RAR) agonists in diabetes and fatty liver disease. METHODS Synthetic agonists for RARβ2 were administered to wild-type (wt) mice in a model of high-fat-diet (HFD)-induced type 2 diabetes (T2D) and to ob/ob and db/db mice (genetic models of obesity-associated T2D). RESULTS We show that administration of synthetic agonists for RARβ2 to either wt mice in a model of HFD-induced T2D or to ob/ob and db/db mice reduces hyperglycaemia, peripheral insulin resistance and body weight. Furthermore, RARβ2 agonists dramatically reduce steatosis, lipid peroxidation and oxidative stress in the liver, pancreas and kidneys of obese, diabetic mice. RARβ2 agonists also lower levels of mRNAs involved in lipogenesis, such as sterol regulatory element-binding transcription factor 1 (SREBP1) and fatty acid synthase, and increase mRNAs that mediate mitochondrial fatty acid β-oxidation, such as CPT1α, in these organs. RARβ2 agonists lower triglyceride levels in these organs, and in muscle. CONCLUSIONS Collectively, our data show that orally active, rapid-acting, high-affinity pharmacological agonists for RARβ2 improve the diabetic phenotype while reducing lipid levels in key insulin target tissues. We suggest that RARβ2 agonists should be useful drugs for T2D therapy and for treatment of hepatic steatosis.
Collapse
MESH Headings
- Animals
- Benzoates/therapeutic use
- Biphenyl Compounds/therapeutic use
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diet, High-Fat/adverse effects
- Drugs, Investigational/therapeutic use
- Hyperglycemia/prevention & control
- Hypoglycemic Agents/therapeutic use
- Insulin Resistance
- Kidney/drug effects
- Kidney/metabolism
- Kidney/pathology
- Lipid Peroxidation/drug effects
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Male
- Mice, Inbred C57BL
- Mice, Mutant Strains
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Non-alcoholic Fatty Liver Disease/complications
- Non-alcoholic Fatty Liver Disease/prevention & control
- Obesity/complications
- Oxidative Stress/drug effects
- Pancreas/drug effects
- Pancreas/metabolism
- Pancreas/pathology
- Receptors, Retinoic Acid/agonists
- Receptors, Retinoic Acid/metabolism
- Thiazoles/therapeutic use
Collapse
Affiliation(s)
- Steven E. Trasino
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, 1300 York Ave., New York, NY 10065, Tel.: 212-746-6250; Fax: 212-746-8858
| | - Xiao-Han Tang
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, 1300 York Ave., New York, NY 10065, Tel.: 212-746-6250; Fax: 212-746-8858
| | - Jose Jessurun
- Department of Pathology, Weill Cornell Medical College of Cornell University/New York Presbyterian Hospital, 525 East 68th Street New York, NY 10065, Tel.: 212-746-2700; Fax: 212-746-8624
| | - Lorraine J. Gudas
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, 1300 York Ave., New York, NY 10065, Tel.: 212-746-6250; Fax: 212-746-8858
- To whom correspondence should be addressed: Lorraine J. Gudas, Department of Pharmacology, Weill Cornell Medical College of Cornell University, 1300 York Ave., New York, NY 10065. Tel.: 212-746-6250; Fax: 212-746-8858;
| |
Collapse
|
126
|
Abstract
In the current review, we discuss limitations and recent advances in animal models of diabetic nephropathy (DN). As in human disease, genetic factors may determine disease severity with the murine FVB and DBA/2J strains being more susceptible to DN than C57BL/6J mice. On the black and tan, brachyuric (BTBR) background, leptin deficient (ob/ob) mice develop many of the pathological features of human DN. Hypertension synergises with hyperglycemia to promote nephropathy in rodents. Moderately hypertensive endothelial nitric oxide synthase (eNOS(-/-)) deficient diabetic mice develop hyaline arteriosclerosis and nodular glomerulosclerosis and induction of renin-dependent hypertension in diabetic Cyp1a1mRen2 rats mimics moderately severe human DN. In addition, diabetic eNOS(-/-) mice and Cyp1a1mRen2 rats recapitulate many of the molecular pathways activated in the human diabetic kidney. However, no model exhibits all the features of human DN; therefore, researchers should consider biochemical, pathological, and transcriptomic data in selecting the most appropriate model to study their molecules and pathways of interest.
Collapse
Affiliation(s)
- Boris Betz
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, Scotland
- Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Jena, Germany
| | - Bryan R Conway
- Centre for Cardiovascular Science, Queen's Medical Research Centre, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland.
| |
Collapse
|
127
|
|
128
|
Platt TL, Beckett TL, Kohler K, Niedowicz DM, Murphy MP. Obesity, diabetes, and leptin resistance promote tau pathology in a mouse model of disease. Neuroscience 2015; 315:162-74. [PMID: 26701291 DOI: 10.1016/j.neuroscience.2015.12.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 12/04/2015] [Accepted: 12/04/2015] [Indexed: 01/22/2023]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) convey an increased risk for developing dementia. The microtubule-associated protein tau is implicated in neurodegenerative disease by undergoing hyperphosphorylation and aggregation, leading to cytotoxicity and neurodegeneration. Enzymes involved in the regulation of tau phosphorylation, such as GSK3β, are tightly associated with pathways found to be dysregulated in T2DM. We have shown previously that leptin-resistant mice, which develop obesity and a diabetic phenotype, display elevated levels of tau phosphorylation. Here we show cells cultured with leptin, an adipokine shown to have neuroprotective effects, reduces tau phosphorylation. To explore how this mechanism works in vivo we transduced an existing diabetic mouse line (Lepr(db/db)) with a tau mutant (tau(P301L)) via adeno-associated virus (AAV). The resulting phenotype included a striking increase in tau phosphorylation and the number of neurofibrillary tangles (NFTs) found within the hippocampus. We conclude that leptin resistance-induced obesity and diabetes accelerates the development of tau pathology. This model of metabolic dysfunction and tauopathy provides a new system in which to explore the mechanisms underlying the ways in which leptin resistance and diabetes influence development of tau pathology, and may ultimately be related to the development of NFTs.
Collapse
Affiliation(s)
- T L Platt
- Department of Molecular and Cellular Biochemistry, University of Kentucky, United States
| | - T L Beckett
- Sanders Brown Center on Aging, University of Kentucky, United States
| | - K Kohler
- Sanders Brown Center on Aging, University of Kentucky, United States
| | - D M Niedowicz
- Department of Molecular and Cellular Biochemistry, University of Kentucky, United States; Sanders Brown Center on Aging, University of Kentucky, United States
| | - M P Murphy
- Department of Molecular and Cellular Biochemistry, University of Kentucky, United States; Sanders Brown Center on Aging, University of Kentucky, United States.
| |
Collapse
|
129
|
Chesi A, Grant SFA. The Genetics of Pediatric Obesity. Trends Endocrinol Metab 2015; 26:711-721. [PMID: 26439977 PMCID: PMC4673034 DOI: 10.1016/j.tem.2015.08.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 08/20/2015] [Accepted: 08/21/2015] [Indexed: 01/24/2023]
Abstract
Obesity among children and adults has notably escalated over recent decades and represents a global major health problem. We now know that both genetic and environmental factors contribute to its complex etiology. Genome-wide association studies (GWAS) have revealed compelling genetic signals influencing obesity risk in adults. Recent reports for childhood obesity revealed that many adult loci also play a role in the pediatric setting. Childhood GWAS have uncovered novel loci below the detection range in adult studies, suggesting that obesity genes may be more easily uncovered in the pediatric setting. Shedding light on the genetic architecture of childhood obesity will facilitate the prevention and treatment of pediatric cases, and will have fundamental implications for diseases that present later in life.
Collapse
Affiliation(s)
- Alessandra Chesi
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Struan F A Grant
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
130
|
Preliminary Characterization of a Leptin Receptor Knockout Rat Created by CRISPR/Cas9 System. Sci Rep 2015; 5:15942. [PMID: 26537785 PMCID: PMC4633582 DOI: 10.1038/srep15942] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 09/29/2015] [Indexed: 12/28/2022] Open
Abstract
Leptin receptor, which is encoded by the diabetes (db) gene and is highly expressed in the choroid plexus, regulatesenergy homeostasis, the balance between food intake and energy expenditure, fertility and bone mass. Here, using CRISPR/Cas9 technology, we created the leptin receptor knockout rat. Homozygous leptin receptor null rats are characterized by obesity, hyperphagia, hyperglycemia, glucose intolerance, hyperinsulinemia and dyslipidemia. Due to long-term poor glycemic control, the leptin receptor knockout rats also develop some diabetic complications such as pancreatic, hepatic and renal lesions. In addition, the leptin receptor knockout rats show a significant decrease in bone volume and bone mineral density of the femur compared with their wild-type littermates. Our model has rescued some deficiency of the existing rodent models, such as the transient hyperglycemia of db/db mice in the C57BL/6J genetic background and the delayed onset of glucose intolerance in the Zucker rats, and it is proven to be a useful animal model for biomedical and pharmacological research on obesity and diabetes.
Collapse
|
131
|
Obesity Leads to Tissue, but not Serum Vitamin A Deficiency. Sci Rep 2015; 5:15893. [PMID: 26522079 PMCID: PMC4629132 DOI: 10.1038/srep15893] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/06/2015] [Indexed: 12/22/2022] Open
Abstract
Obesity negatively affects multiple metabolic pathways, but little is known about the impact of obesity on vitamin A (VA)[retinol (ROL)], a nutrient that regulates expression of genes in numerous pathways essential for human development and health. We demonstrate that obese mice, generated from a high fat diet (HFD) or by genetic mutations (i.e., ob/ob; db/db), have greatly reduced ROL levels in multiple organs, including liver, lungs, pancreas, and kidneys, even though their diets have adequate VA. However, obese mice exhibit elevated serum VA. Organs from obese mice show impaired VA transcriptional signaling, including reductions in retinoic acid receptor (RARα, RARβ2 and RARγ) mRNAs and lower intracellular ROL binding protein Crbp1 (RBP1) levels in VA-storing stellate cells. Reductions in organ VA signaling in obese mice correlate with increasing adiposity and fatty liver (steatosis), while with weight loss VA levels and signaling normalize. Consistent with our findings in obese mice, we show that increasing severity of fatty liver disease in humans correlates with reductions in hepatic VA, VA transcriptional signaling, and Crbp1 levels in VA storing stellate cells. Thus, obesity causes a “silent” VA deficiency marked by reductions in VA levels and signaling in multiple organs, but not detected by serum VA.
Collapse
|
132
|
Datta N, Lindfors S, Miura N, Saleem MA, Lehtonen S. Overexpression of transcription factor FOXC2 in cultured human podocytes upregulates injury markers and increases motility. Exp Cell Res 2015; 340:32-42. [PMID: 26524507 DOI: 10.1016/j.yexcr.2015.10.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 10/09/2015] [Accepted: 10/29/2015] [Indexed: 01/23/2023]
Abstract
Obesity and diabetes-related kidney diseases associate with renal failure and cardiovascular morbidity, and represent a major health issue worldwide. However, the molecular mechanisms leading to their development remain poorly understood. We observed increased expression of transcription factor FoxC2 in the podocytes of obese Zucker rats that are insulin resistant and albuminuric. We also found that depletion of adiponectin, an adipocyte-derived hormone whose secretion is decreased in obesity, upregulated FOXC2 in differentiated human podocytes in vitro. Overexpression of FOXC2 in cultured human podocytes led to increased nuclear expression of FOXC2 associated with a change of cellular morphology. This was accompanied by upregulation of vimentin, a key mesenchymal marker, and active beta-catenin, associated with podocyte injury. We also observed re-organization of the actin cytoskeleton, disrupted localization of the tight junction protein ZO-1, and increased motility of podocytes overexpressing FOXC2. These data indicate that the expression of FOXC2 in podocytes needs to be tightly regulated, and that its overexpression induces a chain of cellular events leading to podocyte dysfunction. These changes may lead to podocyte detachment and depletion ultimately contributing to albuminuria. We also suggest a novel molecular mechanism linking obesity-induced decrease in adiponectin to podocyte dysfunction via upregulation of FOXC2.
Collapse
Affiliation(s)
- Neeta Datta
- Department of Pathology, University of Helsinki, 00290 Helsinki, Finland
| | - Sonja Lindfors
- Department of Pathology, University of Helsinki, 00290 Helsinki, Finland
| | - Naoyuki Miura
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Moin A Saleem
- Academic and Children's Renal Unit, Dorothy Hodgkin Building, Bristol BS1, United Kingdom
| | - Sanna Lehtonen
- Department of Pathology, University of Helsinki, 00290 Helsinki, Finland.
| |
Collapse
|
133
|
Fan YL, Li XQ. Expression of leptin and its receptor in thyroid carcinoma: distinctive prognostic significance in different subtypes. Clin Endocrinol (Oxf) 2015; 83:261-7. [PMID: 25158596 DOI: 10.1111/cen.12598] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/09/2014] [Accepted: 08/21/2014] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To investigate the potential prognostic significance of leptin and its receptor (Ob-R) in thyroid carcinoma. PATIENTS AND METHODS The study cohort consisted of 173 patients including 93 cases with papillary thyroid carcinoma (PTC), 41 cases with follicular thyroid carcinoma (FTC), 25 cases with medullary thyroid carcinoma (MTC) and 14 cases with anaplastic thyroid carcinoma (ATC). We investigated the correlation between clinicopathological features and leptin or Ob-R. The Kaplan-Meier method was used to analyse the survival rate. RESULTS There was a strong correlation of leptin expression with Ob-R expression in PTC, FTC and ATC. For PTC, leptin expression was strongly correlated with older age, larger tumour size, nodal metastasis and advanced stage. Ob-R was significantly correlated with larger tumour size, nodal metastasis and advanced stage. The 5-year disease-free survival (DFS) rate in patients with positive leptin or its receptor expression was lower than that in patients without expression (with statistical difference). For FTC, patients with positive leptin or Ob-R expression developed no recurrence or metastasis during the follow-up. For MTC, Ob-R was significantly correlated with nodal metastasis and advanced stage (P < 0·05). For ATC, patients with positive Ob-R expression had longer median DFS than those with negative expression (436 ± 185 vs 57 ± 71 days), and the difference in the survival rate was statistically significant (P < 0·05). CONCLUSIONS There was a strong correlation of leptin expression with Ob-R expression in PTC, FTC and ATC. Leptin and Ob-R had negative prognostic significance in PTC, while Ob-R may play a protective role in ATC.
Collapse
MESH Headings
- Adenocarcinoma, Follicular/diagnosis
- Adenocarcinoma, Follicular/metabolism
- Adenocarcinoma, Follicular/mortality
- Carcinoma/diagnosis
- Carcinoma/metabolism
- Carcinoma/mortality
- Carcinoma, Neuroendocrine/diagnosis
- Carcinoma, Neuroendocrine/metabolism
- Carcinoma, Neuroendocrine/mortality
- Carcinoma, Papillary
- Cohort Studies
- Disease-Free Survival
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Kaplan-Meier Estimate
- Leptin/metabolism
- Neoplasm Metastasis
- Neoplasm Recurrence, Local
- Prognosis
- Receptors, Leptin/metabolism
- Recurrence
- Survival Rate
- Thyroid Cancer, Papillary
- Thyroid Carcinoma, Anaplastic/diagnosis
- Thyroid Carcinoma, Anaplastic/metabolism
- Thyroid Carcinoma, Anaplastic/mortality
- Thyroid Neoplasms/diagnosis
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/mortality
- Tissue Array Analysis
Collapse
Affiliation(s)
- Yan-Lan Fan
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Medical Oncology, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, China
| | - Xiao-Qiu Li
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
134
|
Sreenivasan J, Schlenner S, Franckaert D, Dooley J, Liston A. The thymoprotective function of leptin is indirectly mediated via suppression of obesity. Immunology 2015; 146:122-9. [PMID: 26059465 DOI: 10.1111/imm.12488] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/19/2015] [Accepted: 06/01/2015] [Indexed: 12/17/2022] Open
Abstract
Leptin is an adipokine that regulates metabolism and plays an important role as a neuroendocrine hormone. Leptin mediates these functions via the leptin receptor, and deficiency in either leptin or its receptor leads to obesity in humans and mice. Leptin has far reaching effects on the immune system, as observed in obese mice, which display decreased thymic function and increased inflammatory responses. With expression of the leptin receptor on T cells and supporting thymic epithelium, aberrant signalling through the leptin receptor has been thought to be the direct cause of thymic involution in obese mice. Here, we demonstrate that the absence of leptin receptor on either thymic epithelial cells or T cells does not lead to the loss of thymic function, demonstrating that the thymoprotective effect of leptin is mediated by obesity suppression rather than direct signalling to the cellular components of the thymus.
Collapse
Affiliation(s)
- Jayasree Sreenivasan
- VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Susan Schlenner
- VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Dean Franckaert
- VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - James Dooley
- VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Adrian Liston
- VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| |
Collapse
|
135
|
Garcia-Galiano D, Allen SJ, Elias CF. Role of the adipocyte-derived hormone leptin in reproductive control. Horm Mol Biol Clin Investig 2015; 19:141-9. [PMID: 25390022 DOI: 10.1515/hmbci-2014-0017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 04/29/2014] [Indexed: 12/11/2022]
Abstract
Achievement of sexual maturation and maintenance of fertility in adulthood are functions that are sensitive to the metabolic status of the organism, particularly the magnitude of fat reserves. In this sense, the adipocyte-derived hormone, leptin, plays a major role in linking metabolic cues and the control of multiple neuroendocrine axes. The hypothalamus is a key site mediating leptin actions, including those involved in the modulation of the hypothalamus-pituitary-gonads (HPG) axis at different stages of development and in different environmental conditions. In the present review, we provide an update of the role of leptin in reproduction and discuss its interactions with neurons, neurotransmitters and downstream targets of the reproductive axis, with a special emphasis on the actions of leptin in the central nervous system. We hope this review will contribute to the understanding of the mechanisms whereby metabolic signals, especially leptin, influence the reproductive neuroendocrine axis modulating its activity in different nutritional states. Special attention will be given to recent advances in the identification of key hypothalamic sites and signaling pathways relevant to leptin's action in reproductive control.
Collapse
|
136
|
Chowen JA, Argente J. Leptin and the brain. Horm Mol Biol Clin Investig 2015; 7:351-60. [PMID: 25961273 DOI: 10.1515/hmbci.2011.113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 07/13/2011] [Indexed: 02/03/2023]
Abstract
Leptin, which comes from the Greek root leptos meaning thin, has been the focus of intense investigation since its discovery in 1994. This hormone belongs to the cytokine family and is produced by adipocytes and circulates in proportion to fat mass, thus serving as a satiety signal and informing central metabolic control centers as to the status of peripheral energy stores. However, it participates in numerous other functions both peripherally and centrally, as indicated by the wide distribution of its various receptor isoforms. Leptin is involved in brain development, most notably in development of hypothalamic centers that control metabolism, but also in other brain areas. It acts as a nutritional cue to indicate adequacy of energy stores for pubertal development and reproductive capacity. The effects of this hormone on behavior and cognition are less well studied, but it clearly is involved in specific aspects of these physiological phenomena. As obesity is a major health problem in many areas of the world, the search for pharmacological treatments to decrease appetite and increase energy expenditure is intense. Understanding the mechanisms of actions of all physiological effects of this hormone is of great interest in the pursuit of such treatment.
Collapse
|
137
|
Saurus P, Kuusela S, Lehtonen E, Hyvönen ME, Ristola M, Fogarty CL, Tienari J, Lassenius MI, Forsblom C, Lehto M, Saleem MA, Groop PH, Holthöfer H, Lehtonen S. Podocyte apoptosis is prevented by blocking the Toll-like receptor pathway. Cell Death Dis 2015; 6:e1752. [PMID: 25950482 PMCID: PMC4669704 DOI: 10.1038/cddis.2015.125] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 03/30/2015] [Accepted: 03/31/2015] [Indexed: 12/30/2022]
Abstract
High serum lipopolysaccharide (LPS) activity in normoalbuminuric patients with type 1 diabetes (T1D) predicts the progression of diabetic nephropathy (DN), but the mechanisms behind this remain unclear. We observed that treatment of cultured human podocytes with sera from normoalbuminuric T1D patients with high LPS activity downregulated 3-phosphoinositide-dependent kinase-1 (PDK1), an activator of the Akt cell survival pathway, and induced apoptosis. Knockdown of PDK1 in cultured human podocytes inhibited antiapoptotic Akt pathway, stimulated proapoptotic p38 MAPK pathway, and increased apoptosis demonstrating an antiapoptotic role for PDK1 in podocytes. Interestingly, PDK1 was downregulated in the glomeruli of diabetic rats and patients with type 2 diabetes before the onset of proteinuria, further suggesting that reduced expression of PDK1 associates with podocyte injury and development of DN. Treatment of podocytes in vitro and mice in vivo with LPS reduced PDK1 expression and induced apoptosis, which were prevented by inhibiting the Toll-like receptor (TLR) signaling pathway with the immunomodulatory agent GIT27. Our data show that LPS downregulates the cell survival factor PDK1 and induces podocyte apoptosis, and that blocking the TLR pathway with GIT27 may provide a non-nephrotoxic means to prevent the progression of DN.
Collapse
Affiliation(s)
- P Saurus
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - S Kuusela
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - E Lehtonen
- 1] Department of Pathology, University of Helsinki, Helsinki, Finland [2] Laboratory Animal Centre, University of Helsinki, Helsinki, Finland
| | - M E Hyvönen
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - M Ristola
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - C L Fogarty
- 1] Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland [2] Division of Nephrology, Helsinki University Central Hospital, Helsinki, Finland [3] Diabetes and Obesity Research Program, Research Program's Unit, University of Helsinki, Helsinki, Finland
| | - J Tienari
- Department of Pathology, HUSLAB and Helsinki University Hospital, Helsinki and Hyvinkää, Finland
| | - M I Lassenius
- 1] Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland [2] Division of Nephrology, Helsinki University Central Hospital, Helsinki, Finland [3] Diabetes and Obesity Research Program, Research Program's Unit, University of Helsinki, Helsinki, Finland
| | - C Forsblom
- 1] Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland [2] Division of Nephrology, Helsinki University Central Hospital, Helsinki, Finland [3] Diabetes and Obesity Research Program, Research Program's Unit, University of Helsinki, Helsinki, Finland
| | - M Lehto
- 1] Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland [2] Division of Nephrology, Helsinki University Central Hospital, Helsinki, Finland [3] Diabetes and Obesity Research Program, Research Program's Unit, University of Helsinki, Helsinki, Finland
| | - M A Saleem
- Bristol Royal Hospital for Children, University of Bristol, Bristol, UK
| | - P-H Groop
- 1] Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland [2] Division of Nephrology, Helsinki University Central Hospital, Helsinki, Finland [3] Diabetes and Obesity Research Program, Research Program's Unit, University of Helsinki, Helsinki, Finland [4] Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - H Holthöfer
- Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - S Lehtonen
- Department of Pathology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
138
|
Przygodzki T, Talar M, Przygodzka P, Watala C. Inhibition of cyclooxygenase-2 causes a decrease in coronary flow in diabetic mice. The possible role of PGE2 and dysfunctional vasodilation mediated by prostacyclin receptor. J Physiol Biochem 2015; 71:351-8. [PMID: 25940857 DOI: 10.1007/s13105-015-0415-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 04/26/2015] [Indexed: 11/25/2022]
Abstract
Several lines of evidence suggest that cyclooxygenase-2 (COX-2) activity can have a beneficial role in the maintenance of vascular tone of the blood vessels in diabetes. Specifically, the increased production of prostacyclin (PGI2) and prostaglandin E2 (PGE2), mediated by COX-2, has been suggested to compensate for decreased synthesis of nitric oxide (NO). The study investigates whether inhibition of COX-2 may reduce the coronary flow in diabetic animals and may also lead to decreased synthesis of prostaglandins. Mice aged 18-20 weeks were used for the study: those with leptin receptor deficiency (db/db) served as a model of diabetes while heterozygous (db/+) mice served as controls. Coronary flow was measured by the Langendorff method, and prostaglandin synthesis by myocardia was assayed in heart perfusates. COX-2 inhibition was found to reduce basal coronary flow in db/db mice but had no effect in db/+ mice. Secretion of PGE2 was found to be higher in db/db mice, while prostacyclin synthesis did not differ. COX-2 inhibition decreased production of both prostaglandins to similar levels in both groups. The use of ONO-1301, a specific agonist for the prostacyclin receptor revealed that vasodilating responses mediated by the receptor were impaired in db/db mice. The expression levels of the receptor in cardiac tissue did not differ between the groups. It is concluded that the increased COX-2 contribution to vasodilation in diabetic animals appears to be partially a result of increased COX-2-dependent synthesis of PGE2 and also may be caused by impaired vasodilation mediated by the prostacyclin receptor.
Collapse
Affiliation(s)
- Tomasz Przygodzki
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Lodz, 92-215, Poland,
| | | | | | | |
Collapse
|
139
|
Beretta M, Bauer M, Hirsch E. PI3K signaling in the pathogenesis of obesity: The cause and the cure. Adv Biol Regul 2015; 58:1-15. [PMID: 25512233 DOI: 10.1016/j.jbior.2014.11.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 11/21/2014] [Accepted: 11/21/2014] [Indexed: 06/04/2023]
Abstract
With the steady rise in the incidence of obesity and its associated comorbidities, in the last decades research aimed at understanding molecular mechanisms that control body weight has gained new interest. Fat gain is frequently associated with chronic adipose tissue inflammation and with peripheral as well as central metabolic derangements, resulting in an impaired hypothalamic regulation of energy homeostasis. Recent attention has focused on the role of phosphatidylinositol 3-kinase (PI3K) in both immune and metabolic response pathways, being involved in the pathophysiology of obesity and its associated metabolic diseases. In this review, we focus on distinct PI3K isoforms, especially class I PI3Ks, mediating inflammatory cells recruitment to the enlarged fat as well as intracellular responses to key hormonal regulators of fat storage, both in adipocytes and in the central nervous system. This integrated view of PI3K functions may ultimately help to develop new therapeutic interventions for the treatment of obesity.
Collapse
Affiliation(s)
- Martina Beretta
- Molecular Biotechnology Center, University of Torino, Torino, Italy; Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Emilio Hirsch
- Molecular Biotechnology Center, University of Torino, Torino, Italy.
| |
Collapse
|
140
|
Ramírez S, Claret M. Hypothalamic ER stress: A bridge between leptin resistance and obesity. FEBS Lett 2015; 589:1678-87. [PMID: 25913783 DOI: 10.1016/j.febslet.2015.04.025] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 04/14/2015] [Accepted: 04/15/2015] [Indexed: 12/21/2022]
Abstract
The prevalence of obesity has increased worldwide at an alarming rate. However, non-invasive pharmacological treatments remain elusive. Leptin resistance is a general feature of obesity, thus strategies aimed at enhancing the sensitivity to this hormone may constitute an excellent therapeutical approach to counteract current obesity epidemics. Nevertheless, the etiology and neuronal basis of leptin resistance remains an enigma. A recent hypothesis gaining substantial experimental support is that hypothalamic endoplasmic reticulum (ER) stress plays a causal role in the development of leptin resistance and obesity. The objective of this review article is to provide an updated view on current evidence connecting hypothalamic ER stress with leptin resistance. We discuss the experimental findings supporting this hypothesis, as well as the potential causes and underlying mechanisms leading to this metabolic disorder. Understanding these mechanisms may provide key insights into the development of novel intervention approaches.
Collapse
Affiliation(s)
- Sara Ramírez
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Marc Claret
- Diabetes and Obesity Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain
| |
Collapse
|
141
|
Chronic stress aggravates glucose intolerance in leptin receptor-deficient (db/db) mice. GENES AND NUTRITION 2015; 10:458. [PMID: 25791744 DOI: 10.1007/s12263-015-0458-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/05/2015] [Indexed: 12/17/2022]
Abstract
Genetic predisposition and environmental challenges interact to determine individual vulnerability to obesity and type 2 diabetes. We previously established a mouse model of chronic subordination stress-induced hyperphagia, obesity, metabolic like-syndrome and insulin resistance in the presence of a high-fat diet. However, it remains to be established if social stress could also aggravate glucose intolerance in subjects genetically predisposed to develop obesity and type 2 diabetes. To answer this question, we subjected genetically obese mice due to deficiency of the leptin receptor (db/db strain) to chronic subordination stress. Over five weeks, subordination stress in db/db mice led to persistent hyperphagia, hyperglycemia and exacerbated glucose intolerance altogether suggestive of an aggravated disorder when compared to controls. On the contrary, body weight and fat mass were similarly affected in stressed and control mice likely due to the hyperactivity shown by subordinate mice. Stressed db/db mice also showed increased plasma inflammatory markers. Altogether our results suggest that chronic stress can aggravate glucose intolerance but not obesity in genetically predisposed subjects on the basis of a disrupted leptin circuitry.
Collapse
|
142
|
Fernández-Formoso G, Pérez-Sieira S, González-Touceda D, Dieguez C, Tovar S. Leptin, 20 years of searching for glucose homeostasis. Life Sci 2015; 140:4-9. [PMID: 25744051 DOI: 10.1016/j.lfs.2015.02.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 12/31/2022]
Abstract
Leptin was discovered in 1994 (20 years ago). In addition to having well-characterized effects on the regulation of energy homeostasis, leptin clearly also plays a major role in metabolic homeostasis. In fact, leptin plays an important role in the regulation of glucose homeostasis independent of food intake and body weight. The mechanism underlying the modulation of glucose metabolism by leptin is not completely understood, although evidence indicates that the effect occurs at both the central and peripheral levels. In this review, we will focus on the role of leptin in glucose homeostasis at the central level and its role in insulin secretion and in counteracting hormones, such as glucagon, growth hormone, cortisol and catecholamines.
Collapse
Affiliation(s)
- Gabriela Fernández-Formoso
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain
| | - Sonia Pérez-Sieira
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - David González-Touceda
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain
| | - Carlos Dieguez
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Sulay Tovar
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain.
| |
Collapse
|
143
|
Ozcan M, Saatci T, Ayar A, Canpolat S, Kelestimur H. Leptin activates cytosolic calcium responses through protein kinase-C dependent mechanism in immortalized RFamide-related peptide-3 neurons. Brain Res 2015; 1601:8-14. [DOI: 10.1016/j.brainres.2014.12.053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 12/20/2014] [Accepted: 12/29/2014] [Indexed: 10/24/2022]
|
144
|
Brown JA, Woodworth HL, Leinninger GM. To ingest or rest? Specialized roles of lateral hypothalamic area neurons in coordinating energy balance. Front Syst Neurosci 2015; 9:9. [PMID: 25741247 PMCID: PMC4332303 DOI: 10.3389/fnsys.2015.00009] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 01/15/2015] [Indexed: 12/26/2022] Open
Abstract
Survival depends on an organism’s ability to sense nutrient status and accordingly regulate intake and energy expenditure behaviors. Uncoupling of energy sensing and behavior, however, underlies energy balance disorders such as anorexia or obesity. The hypothalamus regulates energy balance, and in particular the lateral hypothalamic area (LHA) is poised to coordinate peripheral cues of energy status and behaviors that impact weight, such as drinking, locomotor behavior, arousal/sleep and autonomic output. There are several populations of LHA neurons that are defined by their neuropeptide content and contribute to energy balance. LHA neurons that express the neuropeptides melanin-concentrating hormone (MCH) or orexins/hypocretins (OX) are best characterized and these neurons play important roles in regulating ingestion, arousal, locomotor behavior and autonomic function via distinct neuronal circuits. Recently, another population of LHA neurons containing the neuropeptide Neurotensin (Nts) has been implicated in coordinating anorectic stimuli and behavior to regulate hydration and energy balance. Understanding the specific roles of MCH, OX and Nts neurons in harmonizing energy sensing and behavior thus has the potential to inform pharmacological strategies to modify behaviors and treat energy balance disorders.
Collapse
Affiliation(s)
- Juliette A Brown
- Department of Pharmacology and Toxicology, Michigan State University East Lansing, MI, USA ; Center for Integrative Toxicology East Lansing, MI, USA
| | | | - Gina M Leinninger
- Center for Integrative Toxicology East Lansing, MI, USA ; Department of Physiology, Michigan State University East Lansing, MI, USA
| |
Collapse
|
145
|
Affiliation(s)
- Rexford S Ahima
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, and the Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
146
|
The systemic milieu as a mediator of dietary influence on stem cell function during ageing. Ageing Res Rev 2015; 19:53-64. [PMID: 25481406 DOI: 10.1016/j.arr.2014.11.004] [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] [Received: 09/03/2014] [Revised: 11/20/2014] [Accepted: 11/26/2014] [Indexed: 01/09/2023]
Abstract
The regenerative decline of organisms during ageing is linked to the reduced proliferative activity, impaired function and exhaustion of tissue-specific stem and progenitor cells. Studies using heterochronic parabiosis, involving the surgical attachment of young and old organisms so that they share a common vascular system, have revealed that the systemic environment has a profound effect on stem cell function. In particular, specific youthful rejuvenating circulatory factors reverse age-related declines in stem cell function, whereas the old milieu contains inhibitory factors that impede stem cell function in young animals. Similarly, the effects of certain dietary interventions, namely calorie restriction, also induce a more youthful cellular and molecular phenotype in ageing stem cells throughout the body. Further to this, there are key molecular pathways involved in translating the availability of nutrients into altered stem cell function, including signalling in the insulin and insulin-like growth factor and mechanistic target of rapamycin (mTOR) pathways. In this review, we discuss the potential role of dietary interventions to promote a more rejuvenating systemic milieu in order to enhance stem cell function and promote healthy ageing.
Collapse
|
147
|
Xu L. Leptin action in the midbrain: From reward to stress. J Chem Neuroanat 2014; 61-62:256-65. [DOI: 10.1016/j.jchemneu.2014.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/13/2014] [Accepted: 06/25/2014] [Indexed: 12/11/2022]
|
148
|
Bless EP, Reddy T, Acharya KD, Beltz BS, Tetel MJ. Oestradiol and diet modulate energy homeostasis and hypothalamic neurogenesis in the adult female mouse. J Neuroendocrinol 2014; 26:805-16. [PMID: 25182179 PMCID: PMC4476296 DOI: 10.1111/jne.12206] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/27/2014] [Accepted: 08/27/2014] [Indexed: 12/19/2022]
Abstract
Leptin and oestradiol have overlapping functions in energy homeostasis and fertility, and receptors for these hormones are localised in the same hypothalamic regions. Although, historically, it was assumed that mammalian adult neurogenesis was confined to the olfactory bulbs and the hippocampus, recent research has found new neurones in the male rodent hypothalamus. Furthermore, some of these new neurones are leptin-sensitive and affected by diet. In the present study, we tested the hypothesis that diet and hormonal status modulate hypothalamic neurogenesis in the adult female mouse. Adult mice were ovariectomised and implanted with capsules containing oestradiol (E2 ) or oil. Within each group, mice were fed a high-fat diet (HFD) or maintained on standard chow (STND). All animals were administered i.c.v. 5-bromo-2'-deoxyuridine (BrdU) for 9 days and sacrificed 34 days later after an injection of leptin to induce phosphorylation of signal transducer of activation and transcription 3 (pSTAT3). Brain tissue was immunohistochemically labelled for BrdU (newly born cells), Hu (neuronal marker) and pSTAT3 (leptin sensitive). Although mice on a HFD became obese, oestradiol protected against obesity. There was a strong interaction between diet and hormone on new cells (BrdU+) in the arcuate, ventromedial hypothalamus and dorsomedial hypothalamus. HFD increased the number of new cells, whereas E2 inhibited this effect. Conversely, E2 increased the number of new cells in mice on a STND diet in all hypothalamic regions studied. Although the total number of new leptin-sensitive neurones (BrdU-Hu-pSTAT3) found in the hypothalamus was low, HFD increased these new cells in the arcuate, whereas E2 attenuated this induction. These results suggest that adult neurogenesis in the hypothalamic neurogenic niche is modulated by diet and hormonal status and is related to energy homeostasis in female mice.
Collapse
Affiliation(s)
- E P Bless
- Neuroscience Program, Wellesley College, Wellesley, MA, USA
| | | | | | | | | |
Collapse
|
149
|
Rossetti C, Halfon O, Boutrel B. Controversies about a common etiology for eating and mood disorders. Front Psychol 2014; 5:1205. [PMID: 25386150 PMCID: PMC4209809 DOI: 10.3389/fpsyg.2014.01205] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 10/06/2014] [Indexed: 12/25/2022] Open
Abstract
Obesity and depression represent a growing health concern worldwide. For many years, basic science and medicine have considered obesity as a metabolic illness, while depression was classified a psychiatric disorder. Despite accumulating evidence suggesting that obesity and depression may share commonalities, the causal link between eating and mood disorders remains to be fully understood. This etiology is highly complex, consisting of multiple environmental and genetic risk factors that interact with each other. In this review, we sought to summarize the preclinical and clinical evidence supporting a common etiology for eating and mood disorders, with a particular emphasis on signaling pathways involved in the maintenance of energy balance and mood stability, among which orexigenic and anorexigenic neuropeptides, metabolic factors, stress responsive hormones, cytokines, and neurotrophic factors.
Collapse
Affiliation(s)
- Clara Rossetti
- Center for Psychiatric Neuroscience, Lausanne University Hospital Lausanne, Switzerland
| | - Olivier Halfon
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital Lausanne, Switzerland
| | - Benjamin Boutrel
- Center for Psychiatric Neuroscience, Lausanne University Hospital Lausanne, Switzerland ; Division of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital Lausanne, Switzerland
| |
Collapse
|
150
|
Abstract
Mammals regulate fat mass so that increases or reductions in adipose tissue mass activate responses that favor return to one's previous weight. A reduction in fat mass activates a system that increases food intake and reduces energy expenditure; conversely, overfeeding and rapid adipose tissue expansion reduces food intake and increases energy expenditure. With the identification of leptin nearly two decades ago, the central circuit that defends against reductions in body fat was revealed. However, the systems that defend against rapid expansion of fat mass remain largely unknown. Here we review the physiology of the overfed state and evidence for a distinct regulatory system, which unlike the leptin-mediated system, we propose primarily measures a functional aspect of adipose tissue and not total mass per se.
Collapse
Affiliation(s)
- Yann Ravussin
- Department of Medicine, Columbia University, 1150 St. Nicholas Ave, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, 1150 St. Nicholas Ave, New York, NY 10032, USA
| | - Rudolph L Leibel
- Naomi Berrie Diabetes Center, Columbia University, 1150 St. Nicholas Ave, New York, NY 10032, USA; Department of Pediatrics, Columbia University, 1150 St. Nicholas Ave, New York, NY 10032, USA
| | - Anthony W Ferrante
- Department of Medicine, Columbia University, 1150 St. Nicholas Ave, New York, NY 10032, USA; Naomi Berrie Diabetes Center, Columbia University, 1150 St. Nicholas Ave, New York, NY 10032, USA.
| |
Collapse
|