Network


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

Hotspot


Dive into the research topics where Alessandro Pocai is active.

Publication


Featured researches published by Alessandro Pocai.


Journal of Clinical Investigation | 2008

Activation of hypothalamic S6 kinase mediates diet-induced hepatic insulin resistance in rats

Hiraku Ono; Alessandro Pocai; Yuhua Wang; Hideyuki Sakoda; Tomoichiro Asano; Jonathan M. Backer; Gary J. Schwartz; Luciano Rossetti

Prolonged activation of p70 S6 kinase (S6K) by insulin and nutrients leads to inhibition of insulin signaling via negative feedback input to the signaling factor IRS-1. Systemic deletion of S6K protects against diet-induced obesity and enhances insulin sensitivity in mice. Herein, we present evidence suggesting that hypothalamic S6K activation is involved in the pathogenesis of diet-induced hepatic insulin resistance. Extending previous findings that insulin suppresses hepatic glucose production (HGP) partly via its effect in the hypothalamus, we report that this effect was blunted by short-term high-fat diet (HFD) feeding, with concomitant suppression of insulin signaling and activation of S6K in the mediobasal hypothalamus (MBH). Constitutive activation of S6K in the MBH mimicked the effect of the HFD in normal chow-fed animals, while suppression of S6K by overexpression of dominant-negative S6K or dominant-negative raptor in the MBH restored the ability of MBH insulin to suppress HGP after HFD feeding. These results suggest that activation of hypothalamic S6K contributes to hepatic insulin resistance in response to short-term nutrient excess.


Molecular and Cellular Biology | 2009

The Irs1 branch of the insulin signaling cascade plays a dominant role in hepatic nutrient homeostasis.

Shaodong Guo; Kyle D. Copps; Xiaocheng Dong; Sunmin Park; Zhiyong Cheng; Alessandro Pocai; Luciano Rossetti; Mini P. Sajan; Robert V. Farese; Morris F. White

ABSTRACT We used a Cre-loxP approach to generate mice with varied expression of hepatic Irs1 and Irs2 to establish the contribution of each protein to hepatic nutrient homeostasis. While nutrient-sensitive transcripts were expressed nearly normally in liver lacking Irs2 (LKO2 mice), these transcripts were significantly dysregulated in liver lacking Irs1 (LKO1 mice) or Irs1 and Irs2 together (DKO mice). Similarly, a set of key gluconeogenic and lipogenic genes was regulated nearly normally by feeding in liver retaining a single Irs1 allele without Irs2 (DKO/1 mice) but was poorly regulated in liver retaining one Irs2 allele without Irs1 (DKO/2 mice). DKO/2 mice, but not DKO/1 mice, also showed impaired glucose tolerance and insulin sensitivity—though both Irs1 and Irs2 were required to suppress hepatic glucose production during hyperinsulinemic-euglycemic clamp. In contrast, either hepatic Irs1 or Irs2 mediated suppression of HGP by intracerebroventricular insulin infusion. After 12 weeks on a high-fat diet, postprandial tyrosine phosphorylation of Irs1 increased in livers of control and LKO2 mice, whereas tyrosine phosphorylation of Irs2 decreased in control and LKO1 mice. Moreover, LKO1 mice—but not LKO2 mice—that were fed a high-fat diet developed postprandial hyperglycemia. We conclude that Irs1 is the principal mediator of hepatic insulin action that maintains glucose homeostasis.


Obesity | 2012

The Glucagon Receptor Is Involved in Mediating the Body Weight-Lowering Effects of Oxyntomodulin

Jennifer R. Kosinski; James Hubert; Paul E. Carrington; Gary G. Chicchi; James Mu; Corey Miller; Jin Cao; Elisabetta Bianchi; Antonello Pessi; Ranabir SinhaRoy; Donald J. Marsh; Alessandro Pocai

Oxyntomodulin (OXM) is a peptide secreted postprandially from the L‐cells of the gut that has a weak affinity for both the glucagon‐like peptide‐1 receptor (GLP1R) and the glucagon receptor (GCGR). Peripheral administration of OXM in humans and rodents causes weight loss reducing food intake and increasing energy expenditure. It has been suggested that OXM modulates energy intake solely through GLP1R agonism. Because glucagon decreases food intake in rodents and humans, we examined whether activation of the GCGR is involved in the body weight‐lowering effects of OXM. We identified an equipotent GLP1R‐selective peptide agonist that differs from OXM by only one residue (Q3→E, OXMQ3E), but has no significant GCGR agonist activity in vitro and ∼100‐fold reduced ability to stimulate liver glycogenolysis. Chronic treatment of obese mice with OXM and OXMQ3E demonstrated that OXM exhibits superior weight loss and lipid‐lowering efficacy, and antihyperglycemic activity that is comparable to the corresponding GLP1R‐selective agonist. Studies in Glp1r−/− mice and coadministration of OXM and a GCGR antagonist revealed that the antiobesity effect of OXM requires activation of both GLP1R and GCGR. Our data provide new insight into the mechanism of action of OXM and suggest that activation of GCGR is involved in the body weight‐lowering action of OXM.


Journal of Endocrinology | 2012

Unraveling oxyntomodulin, GLP1's enigmatic brother

Alessandro Pocai

Oxyntomodulin (OXM) is a peptide secreted from the L cells of the gut following nutrient ingestion. OXM is a dual agonist of the glucagon-like peptide-1 receptor (GLP1R) and the glucagon receptor (GCGR) combining the effects of GLP1 and glucagon to act as a potentially more effective treatment for obesity than GLP1R agonists. Injections of OXM in humans cause a significant reduction in weight and appetite, as well as an increase in energy expenditure. Activation of GCGR is classically associated with an elevation in glucose levels, which would be deleterious in patients with T2DM, but the antidiabetic properties of GLP1R agonism would be expected to counteract this effect. Indeed, OXM administration improved glucose tolerance in diet-induced obese mice. Thus, dual agonists of the GCGR and GLP1R represent a new therapeutic approach for diabetes and obesity with the potential for enhanced weight loss and improvement in glycemic control beyond those of GLP1R agonists.


Biopolymers | 2012

Optimization of co‐agonism at GLP‐1 and glucagon receptors to safely maximize weight reduction in DIO‐rodents

Jonathan Day; Vasily Gelfanov; David L. Smiley; Paul E. Carrington; George J. Eiermann; Gary G. Chicchi; Mark D. Erion; Jas Gidda; Nancy A. Thornberry; Matthias H. Tschöp; Donald J. Marsh; Ranabir SinhaRoy; Richard D. DiMarchi; Alessandro Pocai

The ratio of GLP‐1/glucagon receptor (GLP1R/GCGR) co‐agonism that achieves maximal weight loss without evidence of hyperglycemia was determined in diet‐induced obese (DIO) mice chronically treated with GLP1R/GCGR co‐agonist peptides differing in their relative receptor agonism. Using glucagon‐based peptides, a spectrum of receptor selectivity was achieved by a combination of selective incorporation of GLP‐1 sequences, C‐terminal modification, backbone lactam stapling to stabilize helical structure, and unnatural amino acid substitutions at the N‐terminal dipeptide. In addition to α‐amino‐isobutyric acid (Aib) substitution at position two, we show that α,α′‐dimethyl imidazole acetic acid (Dmia) can serve as a potent replacement for the highly conserved histidine at position one. Selective site‐specific pegylation was used to further minimize enzymatic degradation and provide uniform, extended in vivo duration of action. Maximal weight loss devoid of any sign of hyperglycemia was achieved with a co‐agonist comparably balanced for in vitro potency at murine GLP1R and GCGR. This peptide exhibited superior weight loss and glucose lowering compared to a structurally matched pure GLP1R agonist, and to co‐agonists of relatively reduced GCGR tone. Any further enhancement of the relative GCGR agonist potency yielded increased weight loss but at the expense of elevated blood glucose. We conclude that GCGR agonism concomitant with GLP1R agonism constitutes a promising approach to treatment of the metabolic syndrome. However, the relative ratio of GLP1R/GCGR co‐agonism needs to be carefully chosen for each species to maximize weight loss efficacy and minimize hyperglycemia.


Journal of Peptide Science | 2011

DPP‐IV‐resistant, long‐acting oxyntomodulin derivatives

Alessia Santoprete; Elena Capito; Paul E. Carrington; Alessandro Pocai; Marco Finotto; Annunziata Langella; Paolo Ingallinella; Karolina Zytko; Simone Bufali; Simona Cianetti; Maria Veneziano; Fabio Bonelli; Lan Zhu; Edith Monteagudo; Donald J. Marsh; Ranabir SinhaRoy; Elisabetta Bianchi; Antonello Pessi

Obesity is one of the major risk factors for type 2 diabetes, and the development of agents, that can simultaneously achieve glucose control and weight loss, is being actively pursued. Therapies based on peptide mimetics of the gut hormone glucagon‐like peptide 1 (GLP‐1) are rapidly gaining favor, due to their ability to increase insulin secretion in a strictly glucose‐dependent manner, with little or no risk of hypoglycemia, and to their additional benefit of causing a modest, but durable weight loss. Oxyntomodulin (OXM), a 37‐amino acid peptide hormone of the glucagon (GCG) family with dual agonistic activity on both the GLP‐1 (GLP1R) and the GCG (GCGR) receptors, has been shown to reduce food intake and body weight in humans, with a lower incidence of treatment‐associated nausea than GLP‐1 mimetics. As for other peptide hormones, its clinical application is limited by the short circulatory half‐life, a major component of which is cleavage by the enzyme dipeptidyl peptidase IV (DPP‐IV). SAR studies on OXM, described herein, led to the identification of molecules resistant to DPP‐IV degradation, with increased potency as compared to the natural hormone. Analogs derivatized with a cholesterol moiety display increased duration of action in vivo. Moreover, we identified a single substitution which can change the OXM pharmacological profile from a dual GLP1R/GCGR agonist to a selective GLP1R agonist. The latter finding enabled studies, described in detail in a separate study (Pocai A, Carrington PE, Adams JR, Wright M, Eiermann G, Zhu L, Du X, Petrov A, Lassman ME, Jiang G, Liu F, Miller C, Tota LM, Zhou G, Zhang X, Sountis MM, Santoprete A, Capitò E, Chicchi GG, Thornberry N, Bianchi E, Pessi A, Marsh DJ, SinhaRoy R. Glucagon‐like peptide 1/glucagon receptor dual agonism reverses obesity in mice. Diabetes 2009; 58: 2258–2266), which highlight the potential of GLP1R/GCGR dual agonists as a potentially superior class of therapeutics over the pure GLP1R agonists currently in clinical use. Copyright


Endocrinology | 2011

Effects of peripherally administered neuromedin U on energy and glucose homeostasis.

Andrea M. Peier; Kunal Desai; James Hubert; Xiaobing Du; Liming Yang; Ying Qian; Jennifer R. Kosinski; Joseph M. Metzger; Alessandro Pocai; Andrea R. Nawrocki; Ronald B. Langdon; Donald J. Marsh

Neuromedin U (NMU) is a highly conserved peptide reported to modulate energy homeostasis. Pharmacological studies have shown that centrally administered NMU inhibits food intake, reduces body weight, and increases energy expenditure. NMU-deficient mice develop obesity, whereas transgenic mice overexpressing NMU become lean and hypophagic. Two high-affinity NMU receptors, NMUR1 and NMUR2, have been identified. NMUR1 is found primarily in the periphery and NMUR2 primarily in the brain, where it mediates the anorectic effects of centrally administered NMU. Given the broad expression pattern of NMU, we evaluated whether peripheral administration of NMU has effects on energy homeostasis. We observed that acute and chronic peripheral administration of NMU in rodents dose-dependently reduced food intake and body weight and that these effects required NMUR1. The anorectic effects of NMU appeared to be partly mediated by vagal afferents. NMU treatment also increased core body temperature and metabolic rate in mice, suggesting that peripheral NMU modulates energy expenditure. Additionally, peripheral administration of NMU significantly improved glucose excursion. Collectively, these data suggest that NMU functions as a peripheral regulator of energy and glucose homeostasis and the development of NMUR1 agonists may be an effective treatment for diabetes and obesity.


Journal of Peptide Science | 2014

Development of a neuromedin U–human serum albumin conjugate as a long‐acting candidate for the treatment of obesity and diabetes. Comparison with the PEGylated peptide

Philippe Neuner; Andrea M. Peier; Fabio Talamo; Paolo Ingallinella; Armin Lahm; Gaetano Barbato; Annalise Di Marco; Kunal Desai; Karolina Zytko; Ying Qian; Xiaobing Du; Davide Ricci; Edith Monteagudo; Ralph Laufer; Alessandro Pocai; Elisabetta Bianchi; Donald J. Marsh; Antonello Pessi

Neuromedin U (NMU) is an endogenous peptide implicated in the regulation of feeding, energy homeostasis, and glycemic control, which is being considered for the therapy of obesity and diabetes. A key liability of NMU as a therapeutic is its very short half‐life in vivo. We show here that conjugation of NMU to human serum albumin (HSA) yields a compound with long circulatory half‐life, which maintains full potency at both the peripheral and central NMU receptors. Initial attempts to conjugate NMU via the prevalent strategy of reacting a maleimide derivative of the peptide with the free thiol of Cys34 of HSA met with limited success, because the resulting conjugate was unstable in vivo. Use of a haloacetyl derivative of the peptide led instead to the formation of a metabolically stable conjugate. HSA–NMU displayed long‐lasting, potent anorectic, and glucose‐normalizing activity. When compared side by side with a previously described PEG conjugate, HSA–NMU proved superior on a molar basis. Collectively, our results reinforce the notion that NMU‐based therapeutics are promising candidates for the treatment of obesity and diabetes. Copyright


Bioorganic & Medicinal Chemistry | 2013

A PEGylated analog of the gut hormone oxyntomodulin with long-lasting antihyperglycemic, insulinotropic and anorexigenic activity.

Elisabetta Bianchi; Paul E. Carrington; Paolo Ingallinella; Marco Finotto; Alessia Santoprete; Aleksandr Petrov; George J. Eiermann; Jennifer R. Kosinski; Donald J. Marsh; Alessandro Pocai; Ranabir SinhaRoy; Antonello Pessi


Journal of the American Society for Mass Spectrometry | 2014

Quantification of Intact and Truncated Stromal Cell-Derived Factor-1α in Circulation by Immunoaffinity Enrichment and Tandem Mass Spectrometry

Weixun Wang; Bernard K. Choi; Wenyu Li; Zhege Lao; Anita Y. H. Lee; Sandra C. Souza; Nathan A. Yates; Timothy Kowalski; Alessandro Pocai; Lucinda H. Cohen

Collaboration


Dive into the Alessandro Pocai's collaboration.

Researchain Logo
Decentralizing Knowledge