Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Eduardo A. Nillni is active.

Publication


Featured researches published by Eduardo A. Nillni.


Neuron | 2003

The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis.

Michael Cowley; Roy G. Smith; Sabrina Diano; Matthias H. Tschöp; Nina Pronchuk; Kevin L. Grove; Christian J. Strasburger; Martin Bidlingmaier; Mark L. Heiman; Luis Miguel Garcia-Segura; Eduardo A. Nillni; Pablo Mendez; Malcolm J. Low; Peter Sotonyi; Jeffrey M. Friedman; Hongyan Liu; Shirly Pinto; William F. Colmers; Roger D. Cone; Tamas L. Horvath

The gastrointestinal peptide hormone ghrelin stimulates appetite in rodents and humans via hypothalamic actions. We discovered expression of ghrelin in a previously uncharacterized group of neurons adjacent to the third ventricle between the dorsal, ventral, paraventricular, and arcuate hypothalamic nuclei. These neurons send efferents onto key hypothalamic circuits, including those producing neuropeptide Y (NPY), Agouti-related protein (AGRP), proopiomelanocortin (POMC) products, and corticotropin-releasing hormone (CRH). Within the hypothalamus, ghrelin bound mostly on presynaptic terminals of NPY neurons. Using electrophysiological recordings, we found that ghrelin stimulated the activity of arcuate NPY neurons and mimicked the effect of NPY in the paraventricular nucleus of the hypothalamus (PVH). We propose that at these sites, release of ghrelin may stimulate the release of orexigenic peptides and neurotransmitters, thus representing a novel regulatory circuit controlling energy homeostasis.


Journal of Clinical Investigation | 2001

Transcriptional regulation of the thyrotropin-releasing hormone gene by leptin and melanocortin signaling

Mark Harris; Carl J. Aschkenasi; Carol F. Elias; Annie Chandrankunnel; Eduardo A. Nillni; Christian Bjørbæk; Joel K. Elmquist; Jeffrey S. Flier; Anthony N. Hollenberg

Starvation causes a rapid reduction in thyroid hormone levels in rodents. This adaptive response is caused by a reduction in thyrotropin-releasing hormone (TRH) expression that can be reversed by the administration of leptin. Here we examined hypothalamic signaling pathways engaged by leptin to upregulate TRH gene expression. As assessed by leptin-induced expression of suppressor of cytokine signaling-3 (SOCS-3) in fasted rats, TRH neurons in the paraventricular nucleus are activated directly by leptin. To a greater degree, they also contain melanocortin-4 receptors (MC4Rs), implying that leptin can act directly or indirectly by increasing the production of the MC4R ligand, alpha-melanocyte stimulating hormone (alpha-MSH), to regulate TRH expression. We further demonstrate that both pathways converge on the TRH promoter. The melanocortin system activates the TRH promoter through the phosphorylation and DNA binding of the cAMP response element binding protein (CREB), and leptin signaling directly regulates the TRH promoter through the phosphorylation of signal transducer and activator of transcription 3 (Stat3). Indeed, a novel Stat-response element in the TRH promoter is necessary for leptins effect. Thus, the TRH promoter is an ideal target for further characterizing the integration of transcriptional pathways through which leptin acts.


Journal of Biological Chemistry | 2000

Leptin Regulates Prothyrotropin-releasing Hormone Biosynthesis EVIDENCE FOR DIRECT AND INDIRECT PATHWAYS

Eduardo A. Nillni; Charles A. Vaslet; Mark Harris; Anthony N. Hollenberg; Christian Bjørbæk; Jeffrey S. Flier

The hypothalamic-pituitary-thyroid axis is down-regulated during starvation, and falling levels of leptin are a critical signal for this adaptation, acting to suppress preprothyrotropin-releasing hormone (prepro-TRH) mRNA expression in the paraventricular nucleus of the hypothalamus. This study addresses the mechanism for this regulation, using primary cultures of fetal rat hypothalamic neurons as a model system. Leptin dose-dependently stimulated a 10-fold increase in pro-TRH biosynthesis, with a maximum response at 10 nm. TRH release was quantified using immunoprecipitation, followed by isoelectric focusing gel electrophoresis and specific TRH radioimmunoassay. Leptin stimulated TRH release by 7-fold. Immunocytochemistry revealed that a substantial population of cells expressed TRH or leptin receptors and that 8–13% of those expressing leptin receptors coexpressed TRH. Leptin produced a 5-fold induction of luciferase activity in CV-1 cells transfected with a TRH promoter and the long form of the leptin receptor cDNA. Although the above data are consistent with a direct ability of leptin to promote TRH biosynthesis through actions on TRH neurons, addition of α-melanocyte-stimulating hormone produced a 3.5-fold increase in TRH biosynthesis and release, whereas neuropeptide Y treatment suppressed pro-TRH biosynthesis ∼3-fold. Furthermore, the melanocortin-4 receptor antagonist SHU9119 partially inhibited leptin-stimulated TRH release from the neuronal culture. Consequently, our data suggest that leptin regulates the TRH neurons through both direct and indirect pathways.


PLOS ONE | 2009

Hypothalamic Sirt1 regulates food intake in a rodent model system.

Isin Çakir; Mario Perello; Omar Lansari; Norma J. Messier; Charles A. Vaslet; Eduardo A. Nillni

Sirt1 is an evolutionarily conserved NAD+ dependent deacetylase involved in a wide range of processes including cellular differentiation, apoptosis, as well as metabolism, and aging. In this study, we investigated the role of hypothalamic Sirt1 in energy balance. Pharmacological inhibition or siRNA mediated knock down of hypothalamic Sirt1 showed to decrease food intake and body weight gain. Central administration of a specific melanocortin antagonist, SHU9119, reversed the anorectic effect of hypothalamic Sirt1 inhibition, suggesting that Sirt1 regulates food intake through the central melanocortin signaling. We also showed that fasting increases hypothalamic Sirt1 expression and decreases FoxO1 (Forkhead transcription factor) acetylation suggesting that Sirt1 regulates the central melanocortin system in a FoxO1 dependent manner. In addition, hypothalamic Sirt1 showed to regulate S6K signaling such that inhibition of the fasting induced Sirt1 activity results in up-regulation of the S6K pathway. Thus, this is the first study providing a novel role for the hypothalamic Sirt1 in the regulation of food intake and body weight. Given the role of Sirt1 in several peripheral tissues and hypothalamus, potential therapies centered on Sirt1 regulation might provide promising therapies in the treatment of metabolic diseases including obesity.


Cell Metabolism | 2010

SIRT1 deacetylase in POMC neurons is required for homeostatic defenses against diet-induced obesity

Giorgio Ramadori; Teppei Fujikawa; Makoto Fukuda; Jason G. Anderson; Donald A. Morgan; Raul Mostoslavsky; Ronald C. Stuart; Mario Perello; Claudia R. Vianna; Eduardo A. Nillni; Kamal Rahmouni; Roberto Coppari

Feeding on high-calorie (HC) diets induces serious metabolic imbalances, including obesity. Understanding the mechanisms against excessive body weight gain is critical for developing effective antiobesity strategies. Here we show that lack of nicotinamide adenine dinucleotide (NAD(+))-dependent deacetylase SIRT1 in pro-opiomelanocortin (POMC) neurons causes hypersensitivity to diet-induced obesity due to reduced energy expenditure. The ability of leptin to properly engage the phosphoinositide 3-kinase (PI3K) signaling in POMC neurons and elicit remodeling of perigonadal white adipose tissue (WAT) is severely compromised in mutant mice. Also, electrophysiological and histomorphomolecular analyses indicate a selective reduction in sympathetic nerve activity and brown-fat-like characteristics in perigonadal WAT of mutant mice, suggesting a physiologically important role for POMC neurons in controlling this visceral fat depot. In summary, our results provide direct genetic evidence that SIRT1 in POMC neurons is required for normal autonomic adaptations against diet-induced obesity.


Frontiers in Neuroendocrinology | 2010

Regulation of the hypothalamic Thyrotropin Releasing Hormone (TRH) neuron by neuronal and peripheral inputs

Eduardo A. Nillni

The hypothalamic-pituitary-thyroid (HPT) axis plays a critical role in mediating changes in metabolism and thermogenesis. Thus, the central regulation of the thyroid axis by Thyrotropin Releasing Hormone (TRH) neurons in the paraventricular nucleus of the hypothalamus (PVN) is of key importance for the normal function of the axis under different physiological conditions including cold stress and changes in nutritional status. Before the TRH peptide becomes biologically active, a series of tightly regulated processes occur including the proper folding of the prohormone for targeting to the secretory pathway, its post-translational processing, and targeting of the processed peptides to the secretory granules near the plasma membrane of the cell ready for secretion. Multiple inputs coming from the periphery or from neurons present in different areas of the brain including the hypothalamus are responsible for the activation or inhibition of the TRH neuron and in turn affect the output of TRH and the set point of the axis.


Journal of Clinical Investigation | 2004

Regulation of hypothalamic prohormone convertases 1 and 2 and effects on processing of prothyrotropin-releasing hormone

Vanesa C. Sanchez; Jorge Goldstein; Ronald C. Stuart; Virginia Hovanesian; Lihong Huo; Heike Münzberg; Theodore C. Friedman; Christian Bjørbæk; Eduardo A. Nillni

Regulation of energy balance by leptin involves regulation of several neuropeptides, including thyrotropin-releasing hormone (TRH). Synthesized from a larger inactive precursor, its maturation requires proteolytic cleavage by prohormone convertases 1 and 2 (PC1 and PC2). Since this maturation in response to leptin requires prohormone processing, we hypothesized that leptin might regulate hypothalamic PC1 and PC2 expression, ultimately leading to coordinated processing of prohormones into mature peptides. Using hypothalamic neurons, we found that leptin stimulated PC1 and PC2 mRNA and protein expression and also increased PC1 and PC2 promoter activities in transfected 293T cells. Starvation of rats, leading to low serum leptin levels, decreased PC1 and PC2 gene and protein expression in the paraventricular nucleus (PVN) of the hypothalamus. Exogenous administration of leptin to fasted animals restored PC1 levels in the median eminence (ME) and the PVN to approximately the level found in fed control animals. Consistent with this regulation of PCs in the PVN, concentrations of TRH in the PVN and ME were substantially reduced in the fasted animals relative to the fed animals, and leptin reversed this decrease. Further analysis showed that proteolytic cleavage of pro-thyrotropin-releasing hormone (proTRH) at known PC cleavage sites was reduced by fasting and increased in animals given leptin. Combined, these findings suggest that leptin-dependent stimulation of hypothalamic TRH expression involves both activation of trh transcription and stimulation of PC1 and PC2 expression, which lead to enhanced processing of proTRH into mature TRH.


Journal of Biological Chemistry | 1997

Processing of Prothyrotropin-releasing Hormone by the Family of Prohormone Convertases

Philip Schaner; Roberta B. Todd; Nabil G. Seidah; Eduardo A. Nillni

The post-translational processing of prothyrotropin-releasing hormone (pro-TRH25–255) has been extensively studied in our laboratory, and the processing pathway to mature TRH has been elucidated. We have also demonstrated that recombinant PC1 and PC2 process partially purified pro-TRH to cryptic peptides in vitro and that pro-TRH and PC1 mRNAs are coexpressed in primary cultures of hypothalamic neurons. To further define the role of each convertase, and particularly PC1 and PC2, in pro-TRH processing, recombinant vaccinia viruses were used to coexpress the prohormone convertases PC1, PC2, PACE4, PC5-B, furin, or control dynorphin together with rat prepro-TRH in constitutively secreting LoVo cells or in the regulated endocrine GH4C1 cell line. Radioimmunoassays from LoVo-derived secreted products indicated that furin cleaves the precursor to generate both N- and C-terminal intermediates. PC1, PC2, and PACE4 only produced N-terminal intermediates, but less efficiently than furin. In GH4C1 cells, PC1, PC2, furin, PC5-B, and PACE4 produced both N-terminal and C-terminal forms. Significantly, TRH-Gly and TRH were mostly produced by PC1, PC2, and furin. Utilizing gel electrophoresis to further analyze the cleavage specificities of PC1 and PC2, we found that PC1 seems primarily responsible for cleavage to both intermediates and mature TRH, since it generated all products at significantly higher levels than PC2. The addition of 7B2 to the coinfection did not augment the ability of PC2 to cleave pro-TRH to either N- or C-terminal forms.


Journal of Neurochemistry | 2002

Pro‐Thyrotropin‐Releasing Hormone Processing by Recombinant PC1

Eduardo A. Nillni; Theodore C. Friedman; Roberta B. Todd; Nigel P. Birch; Y. Peng Loh; Ivor M.D. Jackson

Abstract: Pro‐thyrotropin‐releasing hormone (proTRH) is the precursor to thyrotropin‐releasing hormone (TRH; pGlu‐His‐Pro‐NH2), the hypothalamic releasing factor that stimulates synthesis and release of thyrotropin from the pituitary gland. Five copies of the TRH progenitor sequence (Gln‐His‐Pro‐Gly) and seven cryptic peptides are formed following posttranslational proteolytic cleavage of the 26‐kDa rat proTRH precursor. The endopeptidase(s) responsible for the physiological conversion of proTRH to the TRH progenitor form is currently unknown. We examined the in vitro processing of [3H]leucine‐labeled or unlabeled proTRH by partially purified recombinant PC1. Recombinant PC1 processed the 26‐kDa TRH precursor by initially cleaving the prohormone after the basic amino acid at either position 153 or 159. Based on the use of our well‐established antibodies, we propose that the initial cleavage gave rise to the formation of a 15‐kDa N‐terminal peptide (preproTRH25–152 or preproTRH25–158) and a 10‐kDa C‐terminal peptide (preproTRH154–255 or preproTRH160–255). Some initial cleavage occurred after amino acid 108 to generate a 16.5‐kDa C‐terminal peptide. The 15‐kDa N‐terminal intermediate was further processed to a 6‐kDa peptide (preproTRH25–76 or preproTRH25–82) and a 3.8‐kDa peptide (preproTRH83–108), whereas the 10‐kDa C‐terminal intermediate was processed to a 5.4‐kDa peptide (preproTRH206–255). The optimal pH for these cleavages was 5.5. ZnCl2, EDTA, EGTA, and the omission of Ca2+ inhibited the formation of pYE27 (preproTRH25–50), one of the proTRH N‐terminal products, by 48, 82, 72, and 45%, respectively. This study provides evidence, for the first time, that recombinant PC 1 enzyme can process proTRH to its predicted peptide intermediates.


Journal of Biological Chemistry | 2013

Obesity Induces Hypothalamic Endoplasmic Reticulum Stress and Impairs Proopiomelanocortin (POMC) Post-translational Processing

Isin Çakir; Nicole E. Cyr; Mario Perello; Bogdan Patedakis Litvinov; Amparo Romero; Ronald C. Stuart; Eduardo A. Nillni

Background: The α-MSH peptide is essential in regulating food intake and energy expenditure. Results: ER stress induced by obesity reduces α-MSH, accumulates POMC, and decreases the enzyme PC2. Conclusion: There is a direct link between obesity and ER stress, resulting in altered POMC processing. Significance: These studies bring a new perspective to how ER stress can regulate energy balance by altering POMC processing. It was shown previously that abnormal prohormone processing or inactive proconverting enzymes that are responsible for this processing cause profound obesity. Our laboratory demonstrated earlier that in the diet-induced obesity (DIO) state, the appetite-suppressing neuropeptide α-melanocyte-stimulating hormone (α-MSH) is reduced, yet the mRNA of its precursor protein proopiomelanocortin (POMC) remained unaltered. It was also shown that the DIO condition promotes the development of endoplasmic reticulum (ER) stress and leptin resistance. In the current study, using an in vivo model combined with in vitro experiments, we demonstrate that obesity-induced ER stress obstructs the post-translational processing of POMC by decreasing proconverting enzyme 2, which catalyzes the conversion of adrenocorticotropin to α-MSH, thereby decreasing α-MSH peptide production. This novel mechanism of ER stress affecting POMC processing in DIO highlights the importance of ER stress in regulating central energy balance in obesity.

Collaboration


Dive into the Eduardo A. Nillni's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mario Perello

National Scientific and Technical Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony N. Hollenberg

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Christian Bjørbæk

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Theodore C. Friedman

Charles R. Drew University of Medicine and Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heike Münzberg

Pennington Biomedical Research Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge