Skip to main content
Top
Published in: Clinical and Experimental Medicine 2/2014

01-05-2014 | Review Article

A ticking clock links metabolic pathways and organ systems function in health and disease

Authors: Manlio Vinciguerra, Maria Florencia Tevy, Gianluigi Mazzoccoli

Published in: Clinical and Experimental Medicine | Issue 2/2014

Login to get access

Abstract

Rhythmic variations with 24-h periodicity hallmark homeostatic regulation, metabolic processes and organ systems function, driven by a circadian timing system composed of central and peripheral oscillators. Recent reports suggest that disrupted circadian rhythmicity of physiology and behavior severely alters body homeostasis. Nuclear receptors and transcriptional regulators sense hormonal and metabolic cues and manage the rhythmic patterns of chromatin remodelling and gene expression, playing a key role in the cross talk between the circadian clock circuitry, the metabolic pathways and the organ systems. The alteration of this cross talk contributes to the pathophysiology of metabolic, degenerative, immune-related and neoplastic diseases.
Literature
2.
go back to reference Gooley JJ, Rajaratnam SM, Brainard GC, Kronauer RE, Czeisler CA, Lockley SW (2010) Spectral responses of the human circadian system depend on the irradiance and duration of exposure to light. Sci Transl Med 2(31):31ra33PubMed Gooley JJ, Rajaratnam SM, Brainard GC, Kronauer RE, Czeisler CA, Lockley SW (2010) Spectral responses of the human circadian system depend on the irradiance and duration of exposure to light. Sci Transl Med 2(31):31ra33PubMed
3.
go back to reference Stevens RG, Hansen J, Costa G, Haus E, Kauppinen T, Aronson KJ, Castaño-Vinyals G, Davis S, Frings-Dresen MH, Fritschi L, Kogevinas M, Kogi K, Lie JA, Lowden A, Peplonska B, Pesch B, Pukkala E, Schernhammer E, Travis RC, Vermeulen R, Zheng T, Cogliano V, Straif K (2011) Considerations of circadian impact for defining ‘shift work’ in cancer studies: IARC working group report. Occup Environ Med 68(2):154–162PubMedCrossRef Stevens RG, Hansen J, Costa G, Haus E, Kauppinen T, Aronson KJ, Castaño-Vinyals G, Davis S, Frings-Dresen MH, Fritschi L, Kogevinas M, Kogi K, Lie JA, Lowden A, Peplonska B, Pesch B, Pukkala E, Schernhammer E, Travis RC, Vermeulen R, Zheng T, Cogliano V, Straif K (2011) Considerations of circadian impact for defining ‘shift work’ in cancer studies: IARC working group report. Occup Environ Med 68(2):154–162PubMedCrossRef
4.
go back to reference Buxton OM, Cain SW, O’Connor SP, Porter JH, Duffy JF, Wang W, Czeisler CA, Shea SA (2012) Adverse metabolic consequences in humans of prolonged sleep restriction combined with circadian disruption. Sci Transl Med 4(129):129ra143 Buxton OM, Cain SW, O’Connor SP, Porter JH, Duffy JF, Wang W, Czeisler CA, Shea SA (2012) Adverse metabolic consequences in humans of prolonged sleep restriction combined with circadian disruption. Sci Transl Med 4(129):129ra143
5.
go back to reference Li J, Lu WQ, Beesley S, Loudon AS, Meng QJ (2012) Lithium impacts on the amplitude and period of the molecular circadian clockwork. PLoS ONE 7(3):e33292PubMedCentralPubMedCrossRef Li J, Lu WQ, Beesley S, Loudon AS, Meng QJ (2012) Lithium impacts on the amplitude and period of the molecular circadian clockwork. PLoS ONE 7(3):e33292PubMedCentralPubMedCrossRef
6.
go back to reference Solt LA, Wang Y, Banerjee S, Hughes T, Kojetin DJ, Lundasen T, Shin Y, Liu J, Cameron MD, Noel R, Yoo SH, Takahashi JS, Butler AA, Kamenecka TM, Burris TP (2012) Regulation of circadian behaviour and metabolism by synthetic REV-ERB agonists. Nature 485(7396):62–68PubMedCentralPubMedCrossRef Solt LA, Wang Y, Banerjee S, Hughes T, Kojetin DJ, Lundasen T, Shin Y, Liu J, Cameron MD, Noel R, Yoo SH, Takahashi JS, Butler AA, Kamenecka TM, Burris TP (2012) Regulation of circadian behaviour and metabolism by synthetic REV-ERB agonists. Nature 485(7396):62–68PubMedCentralPubMedCrossRef
7.
go back to reference Koike N, Yoo SH, Huang HC, Kumar V, Lee C, Kim TK, Takahashi JS (2012) Transcriptional architecture and chromatin landscape of the core circadian clock in mammals. Science 338(6105):349–354PubMedCentralPubMedCrossRef Koike N, Yoo SH, Huang HC, Kumar V, Lee C, Kim TK, Takahashi JS (2012) Transcriptional architecture and chromatin landscape of the core circadian clock in mammals. Science 338(6105):349–354PubMedCentralPubMedCrossRef
8.
go back to reference Cho H, Zhao X, Hatori M, Yu RT, Barish GD, Lam MT, Chong LW, DiTacchio L, Atkins AR, Glass CK, Liddle C, Auwerx J, Downes M, Panda S, Evans RM (2012) Regulation of circadian behaviour and metabolism by REV-ERB-α and REV-ERB-β. Nature 485(7396):123–127PubMedCentralPubMedCrossRef Cho H, Zhao X, Hatori M, Yu RT, Barish GD, Lam MT, Chong LW, DiTacchio L, Atkins AR, Glass CK, Liddle C, Auwerx J, Downes M, Panda S, Evans RM (2012) Regulation of circadian behaviour and metabolism by REV-ERB-α and REV-ERB-β. Nature 485(7396):123–127PubMedCentralPubMedCrossRef
9.
go back to reference Takeda Y, Jothi R, Birault V, Jetten AM (2012) RORγ directly regulates the circadian expression of clock genes and downstream targets in vivo. Nucleic Acids Res 40(17):8519–8535PubMedCentralPubMedCrossRef Takeda Y, Jothi R, Birault V, Jetten AM (2012) RORγ directly regulates the circadian expression of clock genes and downstream targets in vivo. Nucleic Acids Res 40(17):8519–8535PubMedCentralPubMedCrossRef
10.
11.
go back to reference Mazzoccoli G, Pazienza V, Vinciguerra M (2012) Clock genes and clock-controlled genes in the regulation of metabolic rhythms. Chronobiol Int 29(3):227–251PubMedCrossRef Mazzoccoli G, Pazienza V, Vinciguerra M (2012) Clock genes and clock-controlled genes in the regulation of metabolic rhythms. Chronobiol Int 29(3):227–251PubMedCrossRef
12.
go back to reference Scheiermann C, Kunisaki Y, Lucas D, Chow A, Jang JE, Zhang D, Hashimoto D, Merad M, Frenette PS (2012) Adrenergic nerves govern circadian leukocyte recruitment to tissues. Immunity 37(2):290–301PubMedCentralPubMedCrossRef Scheiermann C, Kunisaki Y, Lucas D, Chow A, Jang JE, Zhang D, Hashimoto D, Merad M, Frenette PS (2012) Adrenergic nerves govern circadian leukocyte recruitment to tissues. Immunity 37(2):290–301PubMedCentralPubMedCrossRef
13.
go back to reference Cermakian N, Lange T, Golombek D, Sarkar D, Nakao A, Shibata S, Mazzoccoli G (2013) Cross-talk between the circadian clock circuitry and the immune system. Chronobiol Int (in Press) Cermakian N, Lange T, Golombek D, Sarkar D, Nakao A, Shibata S, Mazzoccoli G (2013) Cross-talk between the circadian clock circuitry and the immune system. Chronobiol Int (in Press)
14.
go back to reference Keller M, Mazuch J, Abraham U, Eom GD, Herzog ED, Volk HD, Kramer A, Maier B (2009) A circadian clock in macrophages controls inflammatory immune responses. Proc Natl Acad Sci USA 106:21407–21412PubMedCentralPubMedCrossRef Keller M, Mazuch J, Abraham U, Eom GD, Herzog ED, Volk HD, Kramer A, Maier B (2009) A circadian clock in macrophages controls inflammatory immune responses. Proc Natl Acad Sci USA 106:21407–21412PubMedCentralPubMedCrossRef
15.
go back to reference Cavadini G, Petrzilka S, Kohler P, Jud C, Tobler I, Birchler T, Fontana A (2007) TNF-alpha suppresses the expression of clock genes by interfering with E-box-mediated transcription. Proc Natl Acad Sci USA 104(31):12843–12848PubMedCentralPubMedCrossRef Cavadini G, Petrzilka S, Kohler P, Jud C, Tobler I, Birchler T, Fontana A (2007) TNF-alpha suppresses the expression of clock genes by interfering with E-box-mediated transcription. Proc Natl Acad Sci USA 104(31):12843–12848PubMedCentralPubMedCrossRef
16.
go back to reference Bollinger T, Leutz A, Leliavski A, Skrum L, Kovac J, Bonacina L, Benedict C, Lange T, Westermann J, Oster H, Solbach W (2011) Circadian clocks in mouse and human CD4 + T cells. PLoS ONE 6:e29801PubMedCentralPubMedCrossRef Bollinger T, Leutz A, Leliavski A, Skrum L, Kovac J, Bonacina L, Benedict C, Lange T, Westermann J, Oster H, Solbach W (2011) Circadian clocks in mouse and human CD4 + T cells. PLoS ONE 6:e29801PubMedCentralPubMedCrossRef
17.
go back to reference Gibbs JE, Blaikley J, Beesley S, Matthews L, Simpson KD, Boyce SH, Farrow SN, Else KJ, Singh D, Ray DW, Loudon AS (2012) The nuclear receptor REV-ERBα mediates circadian regulation of innate immunity through selective regulation of inflammatory cytokines. Proc Natl Acad Sci USA 109:582–587PubMedCentralPubMedCrossRef Gibbs JE, Blaikley J, Beesley S, Matthews L, Simpson KD, Boyce SH, Farrow SN, Else KJ, Singh D, Ray DW, Loudon AS (2012) The nuclear receptor REV-ERBα mediates circadian regulation of innate immunity through selective regulation of inflammatory cytokines. Proc Natl Acad Sci USA 109:582–587PubMedCentralPubMedCrossRef
18.
go back to reference Narasimamurthy R, Hatori M, Nayak SK, Liu F, Panda S, Verma IM (2012) Circadian clock protein cryptochrome regulates the expression of proinflammatory cytokines. Proc Natl Acad Sci USA 109(31):12662–12667PubMedCentralPubMedCrossRef Narasimamurthy R, Hatori M, Nayak SK, Liu F, Panda S, Verma IM (2012) Circadian clock protein cryptochrome regulates the expression of proinflammatory cytokines. Proc Natl Acad Sci USA 109(31):12662–12667PubMedCentralPubMedCrossRef
19.
go back to reference Kaech SM, Cui W (2012) Transcriptional control of effector and memory CD8(+) T cell differentiation. Nat Rev Immunol. doi:10.1038/nri3307. [Epub ahead of print] Kaech SM, Cui W (2012) Transcriptional control of effector and memory CD8(+) T cell differentiation. Nat Rev Immunol. doi:10.​1038/​nri3307. [Epub ahead of print]
20.
go back to reference Ebisawa T, Numazawa K, Shimada H, Izutsu H, Sasaki T, Kato N, Tokunaga K, Mori A, Honma K, Honma S, Shibata S (2010) Self-sustained circadian rhythm in cultured human mononuclear cells isolated from peripheral blood. Neurosci Res 66:223–227PubMedCrossRef Ebisawa T, Numazawa K, Shimada H, Izutsu H, Sasaki T, Kato N, Tokunaga K, Mori A, Honma K, Honma S, Shibata S (2010) Self-sustained circadian rhythm in cultured human mononuclear cells isolated from peripheral blood. Neurosci Res 66:223–227PubMedCrossRef
21.
go back to reference Liu Z, Li Z, Mao K, Zou J, Wang Y, Tao Z, Lin G, Tian L, Ji Y, Wu X, Zhu X, Sun S, Chen W, Xiang C, Sun B (2009) Dec2 promotes Th2 cell differentiation by enhancing IL-2R signaling. J Immunol 183:6320–6329PubMedCrossRef Liu Z, Li Z, Mao K, Zou J, Wang Y, Tao Z, Lin G, Tian L, Ji Y, Wu X, Zhu X, Sun S, Chen W, Xiang C, Sun B (2009) Dec2 promotes Th2 cell differentiation by enhancing IL-2R signaling. J Immunol 183:6320–6329PubMedCrossRef
23.
go back to reference Schmutz I, Ripperger JA, Baeriswyl-Aebischer S, Albrecht U (2010) The mammalian clock component PERIOD2 coordinates circadian output by interaction with nuclear receptors. Genes Dev 24:345–357PubMedCentralPubMedCrossRef Schmutz I, Ripperger JA, Baeriswyl-Aebischer S, Albrecht U (2010) The mammalian clock component PERIOD2 coordinates circadian output by interaction with nuclear receptors. Genes Dev 24:345–357PubMedCentralPubMedCrossRef
24.
go back to reference Silver AC, Arjona A, Walker WE, Fikrig E (2012) The circadian clock controls toll-like receptor 9-mediated innate and adaptive immunity. Immunity 36(2):251–261PubMedCentralPubMedCrossRef Silver AC, Arjona A, Walker WE, Fikrig E (2012) The circadian clock controls toll-like receptor 9-mediated innate and adaptive immunity. Immunity 36(2):251–261PubMedCentralPubMedCrossRef
25.
go back to reference Venteclef N, Jakobsson T, Steffensen KR, Treuter E (2011) Metabolic nuclear receptor signaling and the inflammatory acute phase response. Trends Endocrinol Metab 22:333–343PubMedCrossRef Venteclef N, Jakobsson T, Steffensen KR, Treuter E (2011) Metabolic nuclear receptor signaling and the inflammatory acute phase response. Trends Endocrinol Metab 22:333–343PubMedCrossRef
26.
go back to reference Klotz L, Burgdorf S, Dani I, Saijo K, Flossdorf J, Hucke S, Alferink J, Nowak N, Beyer M, Mayer G, Langhans B, Klockgether T, Waisman A, Eberl G, Schultze J, Famulok M, Kolanus W, Glass C, Kurts C, Knolle PA (2009) The nuclear receptor PPAR gamma selectively inhibits Th17 differentiation in a T cell-intrinsic fashion and suppresses CNS autoimmunity. J Exp Med 206:2079–2089PubMedCentralPubMedCrossRef Klotz L, Burgdorf S, Dani I, Saijo K, Flossdorf J, Hucke S, Alferink J, Nowak N, Beyer M, Mayer G, Langhans B, Klockgether T, Waisman A, Eberl G, Schultze J, Famulok M, Kolanus W, Glass C, Kurts C, Knolle PA (2009) The nuclear receptor PPAR gamma selectively inhibits Th17 differentiation in a T cell-intrinsic fashion and suppresses CNS autoimmunity. J Exp Med 206:2079–2089PubMedCentralPubMedCrossRef
27.
go back to reference Rauen T, Juang YT, Hedrich CM, Kis-Toth K, Tsokos GC (2012) A novel isoform of the orphan receptor RORγt suppresses IL-17 production in human T cells. Genes Immun 13:346–350PubMedCrossRef Rauen T, Juang YT, Hedrich CM, Kis-Toth K, Tsokos GC (2012) A novel isoform of the orphan receptor RORγt suppresses IL-17 production in human T cells. Genes Immun 13:346–350PubMedCrossRef
28.
go back to reference Lardone PJ, Guerrero JM, Fernández-Santos JM, Rubio A, Martín-Lacave I, Carrillo-Vico A (2011) Melatonin synthesized by T lymphocytes as a ligand of the retinoic acid-related orphan receptor. J Pineal Res 51:454–462PubMedCrossRef Lardone PJ, Guerrero JM, Fernández-Santos JM, Rubio A, Martín-Lacave I, Carrillo-Vico A (2011) Melatonin synthesized by T lymphocytes as a ligand of the retinoic acid-related orphan receptor. J Pineal Res 51:454–462PubMedCrossRef
29.
go back to reference Wong SH, Walker JA, Jolin HE, Drynan LF, Hams E, Camelo A, Barlow JL, Neill DR, Panova V, Koch U, Radtke F, Hardman CS, Hwang YY, Fallon PG, McKenzie AN (2012) Transcription factor RORα is critical for nuocyte development. Nat Immunol 13:229–236PubMedCentralPubMedCrossRef Wong SH, Walker JA, Jolin HE, Drynan LF, Hams E, Camelo A, Barlow JL, Neill DR, Panova V, Koch U, Radtke F, Hardman CS, Hwang YY, Fallon PG, McKenzie AN (2012) Transcription factor RORα is critical for nuocyte development. Nat Immunol 13:229–236PubMedCentralPubMedCrossRef
30.
go back to reference Wang NS, McHeyzer-Williams LJ, Okitsu SL, Burris TP, Reiner SL, McHeyzer-Williams MG (2012) Divergent transcriptional programming of class-specific B cell memory by T-bet and RORα. Nat Immunol 13(6):604–611PubMedCentralPubMedCrossRef Wang NS, McHeyzer-Williams LJ, Okitsu SL, Burris TP, Reiner SL, McHeyzer-Williams MG (2012) Divergent transcriptional programming of class-specific B cell memory by T-bet and RORα. Nat Immunol 13(6):604–611PubMedCentralPubMedCrossRef
31.
go back to reference Scheer FA, Hilton MF, Mantzoros CS, Shea SA (2009) Adverse metabolic and cardiovascular consequences of circadian misalignment. Proc Natl Acad Sci USA 106(11):4453–4458PubMedCentralPubMedCrossRef Scheer FA, Hilton MF, Mantzoros CS, Shea SA (2009) Adverse metabolic and cardiovascular consequences of circadian misalignment. Proc Natl Acad Sci USA 106(11):4453–4458PubMedCentralPubMedCrossRef
32.
go back to reference Dijk DJ, Duffy JF, Silva EJ, Shanahan TL, Boivin DB, Czeisler CA (2012) Amplitude reduction and phase shifts of melatonin, cortisol and other circadian rhythms after a gradual advance of sleep and light exposure in humans. PLoS ONE 7(2):e30037PubMedCentralPubMedCrossRef Dijk DJ, Duffy JF, Silva EJ, Shanahan TL, Boivin DB, Czeisler CA (2012) Amplitude reduction and phase shifts of melatonin, cortisol and other circadian rhythms after a gradual advance of sleep and light exposure in humans. PLoS ONE 7(2):e30037PubMedCentralPubMedCrossRef
33.
go back to reference Singh NP, Singh UP, Singh B, Price RL, Nagarkatti M, Nagarkatti PS (2011) Activation of aryl hydrocarbon receptor (AhR) leads to reciprocal epigenetic regulation of FoxP3 and IL-17 expression and amelioration of experimental colitis. PLoS ONE 6(8):e23522PubMedCentralPubMedCrossRef Singh NP, Singh UP, Singh B, Price RL, Nagarkatti M, Nagarkatti PS (2011) Activation of aryl hydrocarbon receptor (AhR) leads to reciprocal epigenetic regulation of FoxP3 and IL-17 expression and amelioration of experimental colitis. PLoS ONE 6(8):e23522PubMedCentralPubMedCrossRef
34.
go back to reference Feng D, Liu T, Sun Z, Bugge A, Mullican SE, Alenghat T, Liu XS, Lazar MA (2011) A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism. Science 331(6022):1315–1319PubMedCentralPubMedCrossRef Feng D, Liu T, Sun Z, Bugge A, Mullican SE, Alenghat T, Liu XS, Lazar MA (2011) A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism. Science 331(6022):1315–1319PubMedCentralPubMedCrossRef
35.
go back to reference Bugge A, Feng D, Everett LJ, Briggs ER, Mullican SE, Wang F, Jager J, Lazar MA (2012) Rev-erbα and Rev-erbβ coordinately protect the circadian clock and normal metabolic function. Genes Dev 26(7):657–667PubMedCentralPubMedCrossRef Bugge A, Feng D, Everett LJ, Briggs ER, Mullican SE, Wang F, Jager J, Lazar MA (2012) Rev-erbα and Rev-erbβ coordinately protect the circadian clock and normal metabolic function. Genes Dev 26(7):657–667PubMedCentralPubMedCrossRef
36.
go back to reference Gillum MP, Kotas ME, Erion DM, Kursawe R, Chatterjee P, Nead KT, Muise ES, Hsiao JJ, Frederick DW, Yonemitsu S, Banks AS, Qiang L, Bhanot S, Olefsky JM, Sears DD, Caprio S, Shulman GI (2011) SirT1 regulates adipose tissue inflammation. Diabetes 60(12):3235–3245PubMedCentralPubMedCrossRef Gillum MP, Kotas ME, Erion DM, Kursawe R, Chatterjee P, Nead KT, Muise ES, Hsiao JJ, Frederick DW, Yonemitsu S, Banks AS, Qiang L, Bhanot S, Olefsky JM, Sears DD, Caprio S, Shulman GI (2011) SirT1 regulates adipose tissue inflammation. Diabetes 60(12):3235–3245PubMedCentralPubMedCrossRef
37.
go back to reference Chalkiadaki A, Guarente L (2012) High-fat diet triggers inflammation-induced cleavage of SIRT1 in adipose tissue to promote metabolic dysfunction. Cell Metab 16(2):180–188PubMedCentralPubMedCrossRef Chalkiadaki A, Guarente L (2012) High-fat diet triggers inflammation-induced cleavage of SIRT1 in adipose tissue to promote metabolic dysfunction. Cell Metab 16(2):180–188PubMedCentralPubMedCrossRef
38.
go back to reference Straub RH, Cutolo M (2007) Circadian rhythms in rheumatoid arthritis: implications for pathophysiology and therapeutic management. Arthritis Rheum 56(2):399–408PubMedCrossRef Straub RH, Cutolo M (2007) Circadian rhythms in rheumatoid arthritis: implications for pathophysiology and therapeutic management. Arthritis Rheum 56(2):399–408PubMedCrossRef
39.
go back to reference Pongratz G, Straub RH (2013) Role of peripheral nerve fibres in acute and chronic inflammation in arthritis. Nat Rev Rheumatol 9(2):117–126PubMedCrossRef Pongratz G, Straub RH (2013) Role of peripheral nerve fibres in acute and chronic inflammation in arthritis. Nat Rev Rheumatol 9(2):117–126PubMedCrossRef
40.
go back to reference Kang JE, Lim MM, Bateman RJ, Lee JJ, Smyth LP, Cirrito JR, Fujiki N, Nishino S, Holtzman DM (2009) Amyloid-beta dynamics are regulated by orexin and the sleep-wake cycle. Science 326(5955):1005–1007PubMedCentralPubMedCrossRef Kang JE, Lim MM, Bateman RJ, Lee JJ, Smyth LP, Cirrito JR, Fujiki N, Nishino S, Holtzman DM (2009) Amyloid-beta dynamics are regulated by orexin and the sleep-wake cycle. Science 326(5955):1005–1007PubMedCentralPubMedCrossRef
41.
go back to reference Roh JH, Huang Y, Bero AW, Kasten T, Stewart FR, Bateman RJ, Holtzman DM (2012) Disruption of the sleep-wake cycle and diurnal fluctuation of β-amyloid in mice with Alzheimer’s disease pathology. Sci Transl Med 4(150):150ra122PubMedCentralPubMed Roh JH, Huang Y, Bero AW, Kasten T, Stewart FR, Bateman RJ, Holtzman DM (2012) Disruption of the sleep-wake cycle and diurnal fluctuation of β-amyloid in mice with Alzheimer’s disease pathology. Sci Transl Med 4(150):150ra122PubMedCentralPubMed
Metadata
Title
A ticking clock links metabolic pathways and organ systems function in health and disease
Authors
Manlio Vinciguerra
Maria Florencia Tevy
Gianluigi Mazzoccoli
Publication date
01-05-2014
Publisher
Springer Milan
Published in
Clinical and Experimental Medicine / Issue 2/2014
Print ISSN: 1591-8890
Electronic ISSN: 1591-9528
DOI
https://doi.org/10.1007/s10238-013-0235-8

Other articles of this Issue 2/2014

Clinical and Experimental Medicine 2/2014 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine