Next Article in Journal
Natural and Synthetic Xanthone Derivatives Counteract Oxidative Stress via Nrf2 Modulation in Inflamed Human Macrophages
Next Article in Special Issue
DGCR8 Microprocessor Subunit Mutation and Expression Deregulation in Thyroid Lesions
Previous Article in Journal
In Vitro and In Silico Analysis of New n-Butyl and Isobutyl Quinoxaline-7-carboxylate 1,4-di-N-oxide Derivatives against Trypanosoma cruzi as Trypanothione Reductase Inhibitors
Previous Article in Special Issue
SOCS3 Ablation in Leptin Receptor-Expressing Cells Causes Autonomic and Cardiac Dysfunctions in Middle-Aged Mice despite Improving Energy and Glucose Metabolism
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Liver Brain Interactions: Focus on FGF21 a Systematic Review

1
Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
2
CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
3
Unidad de Metodología de la Investigación, Fundación Instituto de Investigación de Santiago (FIDIS), 15706 Santiago de Compostela, Spain
4
Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, 5201 Bergen, Norway
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(21), 13318; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232113318
Submission received: 12 September 2022 / Revised: 21 October 2022 / Accepted: 29 October 2022 / Published: 1 November 2022

Abstract

:
Fibroblast growth factor 21 is a pleiotropic hormone secreted mainly by the liver in response to metabolic and nutritional challenges. Physiologically, fibroblast growth factor 21 plays a key role in mediating the metabolic responses to fasting or starvation and acts as an important regulator of energy homeostasis, glucose and lipid metabolism, and insulin sensitivity, in part by its direct action on the central nervous system. Accordingly, pharmacological recombinant fibroblast growth factor 21 therapies have been shown to counteract obesity and its related metabolic disorders in both rodents and nonhuman primates. In this systematic review, we discuss how fibroblast growth factor 21 regulates metabolism and its interactions with the central nervous system. In addition, we also state our vision for possible therapeutic uses of this hepatic-brain axis.

1. Introduction

1.1. Obesity

Changes in diet and physical activity have increased the prevalence of obesity in a relatively short time span [1,2,3,4]. Obesity is defined as an abnormal or excessive accumulation of fat that can be detrimental to health and is caused by excessive nutrient intake over time [5].
The biological mechanisms that regulate food intake were evolutionary adapted in periods where nutrients were scarce and the ability to overeat and store energy when food was present was crucial for survival. This is in stark contrast to our current situation, where food is present in excess in most societies, and our propensity to overeat when food is available, together with the great capacity of our body to store energy in the form of fat, rather represents a great disadvantage to modern humans and gives rise to a range of human health problems [5].
Unlike other common diseases, obesity seems to have an obvious solution that starts with the adjustment of food intake and energy expenditure. However, obesity is a complex and multifactorial disease that is composed of biological, social, and behavioral influences, and treatment by lifestyle changes has been shown to be ineffective [4,6,7]. Therefore, biological and clinical evidence reveals an interaction between genes and the environment that discredits the belief that body weight can be controlled exclusively voluntarily [8].
Obesity is associated with comorbidities such as cardiovascular diseases, type II diabetes mellitus (T2DM), or cancer, among others [9,10]. Therefore, a better understanding of the underlying causes of obesity is urgently required in order to lay the groundwork for the development of new therapeutic strategies [11,12,13].

1.2. Brain Homeostatic Mechanisms and Energy Balance

The Central Nervous System (CNS) plays a fundamental role in the control of the metabolic homeostasis of the organism. Since one of the most important functions for survival is to keep us fed and in good nutritional status, the regulation of the metabolism works through a homeostatic system that balances food intake and energy expenditure. It is a complex system, since it is essential for our survival, and food intake is also controlled by the integration of different cognitive, hedonic, and emotional signals that lead to behavioral, autonomic, and endocrine responses [14,15,16,17]. This maintenance of homeostasis is of great biological importance in order to guarantee a perfect balance between nutrients and energy [16,17,18].
The hypothalamus is a region of the brain composed of nuclei interconnected by axonal projections and is the most studied area in terms of the regulation of food intake and body weight [17,18,19,20]. Its anatomical location, at the base of the brain and adjacent to the median eminence (an organ that receives abundant capillary vascularization), allows it to capture the nutrients and hormones secreted into the bloodstream that send information about the energy state, acting as homeostatic feedback signals in order to maintain this metabolic balance [1,8,21].
The hypothalamus, especially the mediobasal hypothalamus, is an important site of action. Included in this neural network are the arcuate nucleus (ARC) and the ventromedial nucleus (VMH). The ARC nucleus is best positioned to receive signals from the periphery and develop a homeostatic response to peripheral tissues, and is thus considered the “master hypothalamic center” for feeding control [8,19].
In the ARC, we can distinguish two differentiated neuronal populations with antagonistic functions. On the one hand, a neuronal population that expresses neuropeptide Y/Agouti-related peptide (NPY/AgRP) induces a positive energy balance; and on the other hand, neurons that express proopiomelanocortin (POMC), which in turn, induces a negative energy balance. Both types of neurons regulate food intake, energy expenditure, and nutrient partitioning [22,23,24,25].
ARC neurons send projections primarily to other “second-order” neurons located in other hypothalamic nuclei, such as the dorsomedial nucleus, paraventricular nucleus (PVH), or lateral hypothalamic area (LHA) [19,20,21]. These nuclei are responsible for integrating hormonal and nutritional metabolic signals from the peripheral circulation, generating a coordinated response [20,22].
Peripheral signals that regulate metabolic control in the CNS may originate from many organs. However, in recent years, the importance of the liver has gained attention and it is considered a master metabolic organ that integrates peripheral nutrient status that is signaled to the brain.
Therefore, in this review, we discuss how a signal derived from the liver, the peptide fibroblast growth factor 21 (FGF21), controls food intake and energy balance and how some of its metabolic actions are induced by its activity in the CNS.

1.3. Fibroblast Growth Factor21 and Metabolic Control

FGF21 is a pleiotropic hormone that was discovered in the year 2000 [26]. FGF21 is secreted mainly by the liver in response to metabolic and nutritional challenges [27,28], but is also expressed by adipose tissue, skeletal muscle, and the pancreas [29,30,31,32,33]. FGF21 acts through cell-surface receptors comprised of conventional FGF receptors (FGFRs), with tyrosine kinase activity in complex with the single-pass transmembrane protein β-Klotho. These receptors are relatively abundantly expressed, both in peripheral tissues, such as brown adipose tissue (BAT) and white adipose tissue (WAT), as well as in some regions of the CNS, such as the hypothalamus and hindbrain [34,35,36,37,38,39,40,41,42,43].
FGF21 acts as an important regulator of energy homeostasis, glucose and lipid metabolism, and insulin sensitivity (Figure 1).
Physiologically, FGF21 plays a key role in mediating the metabolic responses to fasting or starvation, including fatty acid oxidation and ketogenesis [32,33,44]. FGF21 in the liver may also be induced by low protein and high carbohydrate diets, and it has broad effects on glucose and fatty acid metabolism [34,45]. Interestingly, while low protein diets increase FGF21 in the liver, it has also been reported that they decrease the concentration in the hypothalamus [46]. Importantly, high levels of FGF21 have been associated with different metabolic diseases, such as obesity [47], T2DM [48], non-alcoholic steatohepatitis (NASH) [49], cardiometabolic disorders [50], and congenital or acquired lipodystrophy [51,52]. Interestingly, FGF21 levels in the muscle are associated with mitochondrial inflammatory myopathy, leading to altered myofiber morphology [53]. FGF21 thus serves as a disease marker, but rather than driving disease development, it is believed that the elevated FGF21 levels are not sufficient to counteract disease development. Consistently, pharmacological recombinant FGF21 therapies have been shown to counteract obesity and its related metabolic disorders in both rodents and nonhuman primates [54,55,56,57,58,59]. Furthermore, FGF21 is the downstream target of both peroxisome proliferator-activated receptor-alpha (PPARa) and gamma (PPARg), and a growing body of evidence suggests that the glucose-lowering and insulin-sensitizing effects of the PPARg agonist thiazolidinediones and the therapeutic benefits of the PPARa agonist fenofibrate on lipid profiles are mediated, in part, by FGF21 induction [60,61,62].
While the liver is the predominant site for FGF21 production, adipocytes are suggested to be the main target of FGF21 action [35,63,64] (Figure 1). FGF21 acts directly on adipocytes to stimulate glucose uptake, and adiponectin secretion [55,65,66,67]. In WAT, FGF21 stimulates glucose uptake in an insulin-independent manner [56], modulates lipolysis [68], and potentiates PPARg activity [61]. There is also compelling evidence showing that FGF21 is involved in the thermogenic functions of brown adipocytes [69,70]. In this sense, FGF21 is expressed and secreted in both WAT and BAT [47,69], and the autocrine actions of FGF21 in adipocytes play obligatory roles in mediating the metabolic benefits of PPARg on glucose homeostasis and peripheral insulin sensitivity by forming a feed-forward loop with this nuclear receptor [61]. Moreover, FGF21 injection increased energy expenditure and adiponectin secretion from adipose tissue. Interestingly, the increased energy expenditure by FGF21 administration was attenuated in adiponectin-null mice [65,66]. The requirement of adiponectin for the full effect of FGF21 suggests that adiponectin is important for fatty acid mobilization in WAT under lipolytic conditions. Notably, lipodystrophic mice with reduced adipose tissue are refractory to both acute and chronic effects of systemic FGF21 administration, which is used to decrease blood glucose and increase insulin sensitivity. FGF21 responsiveness was completely restored after the transplantation of WAT into these mice, confirming that adipose tissue is a predominant site contributing to the antidiabetic activities of FGF21 [63]. However, it is currently unclear how FGF21 controls systemic metabolic homeostasis via its actions in adipocytes.

1.4. Central Fibroblast Growth Factor 21 Actions

FGF21 increases energy expenditure and decreases body weight and blood glucose, insulin, and hepatic triglyceride concentrations in rodent models of obesity, in part by acting directly on the nervous system to induce sympathetic outflow to BAT and WAT, promoting in turn thermogenesis and “browning” in these tissues [71,72,73,74] (Figure 2).
In agreement with the pharmacological data, genetic loss of function models of FGF21 aggravates obesity-induced and impairs the thermogenic response, possibly via increased hypothalamic inflammation [75]. Interestingly, by using B-klotho tissue-specific loss-of-function models, the actions of FGF21 on body weight were found to not rely on the ability of FGFs to activate b-klotho in the liver or adipose tissue. In fact, FGF21 required β-klotho-containing receptor complexes in neurons to decrease body weight and circulating glucose and insulin concentrations in diet-induced obese mice [73] (Figure 2).
These data imply that the therapeutic efficacy of FGF-based drugs for treating metabolic diseases relies on their ability to activate b-klotho in the CNS [74]. However, other reports highlight the importance of FGFR1 in the FGF21 action on glucose homeostasis during prolonged fasting. FGF21 acts directly on the hypothalamic neurons to activate the mitogen-activated protein kinase extracellular signal-related kinase 1/2, thereby stimulating the expression of corticotropin-releasing hormone by activation of the transcription factor cAMP response element binding protein in a process dependent on FGFR1 [76].
On the other hand, it has been shown that physiologic, transgenic, and pharmacological activation of FGF21 signaling increases the total caloric intake in rodents [3,56,72,77,78] and fish [79]. Moreover, pharmacological and genetic studies show that FGF21 also induces water intake in part by its actions in the hypothalamic SIM-1 positive neurons [80]. However, as we pointed out before, the genetic or pharmacological activation of FGF21 has the primary effect of increasing thermogenesis and energy expenditure, thereby causing overall weight loss.
In this sense, it is clear that the nervous system plays a crucial role in regulating FGF21’s effect on the intake of liquids or food. FGF21 crosses the blood–brain barrier through simple diffusion [81], and its effects are significantly blunted in animals lacking FGF21-receptors broadly in neurons [78,82,83,84,85].
It is not clear which neuronal mechanisms are behind this orexigenic action of FGF21. Nevertheless, when β-klotho was deleted from vesicular glutamatergic transporter 2 (Vglut2)-expressing neurons, the feeding response to dietary protein was lost [86] (Figure 3). Moreover, FGF21 acts in the ARC to increase the expression of the AgRP and NPY (Figure 3), whereas different studies report about reducing or not affecting cocaine and amphetamine-regulated transcripts or POMC [54,87,88].
Furthermore, some studies show that brain FGF21 signaling is necessary not only for normal food intake but also for macronutrient preference [78,82,83,84]. In this sense, it has been proposed that β-klotho-containing receptor complexes in PVN neurons [82] are part of the neurocircuit mechanisms underlying FGF21’s effect on sweet taste preference (Figure 3). Moreover, deleting β-klotho from Vglut2 neurons, but not gamma-aminobutyric acid (GABA)-ergic or dopaminergic neurons, eliminated the effect of FGF21 on the consumption of sucrose or saccharin [83]. Conversely, genetic activation of β-klotho in Vglut2 neurons decreased sucrose preference [85].
Interestingly, recent studies have shown that FGF21 is produced and secreted in hypothalamic tanycytes [89,90] and activates GABA-containing neurons expressing dopamine receptor 2 (DRd2) in the LHA, that in turn, has been shown to be part of the molecular pathway of the protective effect of prolonged lactation on obesity [90] (Figure 3). Notably, the improvement in body weight due to prolonged breastfeeding in obese mice is due to an increase in the thermogenesis of BAT but not to reduced food intake [90].
In contrast, another study proposes that FGF21 is not expressed in the hypothalamus and, thus, it cannot perform metabolic actions or induce sugar intake in an autocrine manner in this brain area. Instead, it was proposed that FGF21 is exclusively produced from the retrosplenial cortex, where it enhances spatial memory [91].
Nevertheless, accumulating evidence suggests that FGF21 is expressed and mediates its metabolic actions in the brain, and one study links the action of FGF21 and sweet tasting with the mesolimbic dopamine pathway, an area pertaining to the reward system (Figure 4).
Sweets activate ventral tegmental area (VTA) dopamine neurons and increase the dopamine release in the nucleus accumbens (NAc) [87]. Another study supports this neuronal connectivity but with opposite results, and shows that chronic FGF21 administration significantly reduces dopamine and dopamine-related metabolites and increases the expression of the dopamine transporter in the NAc. Moreover, this last study showed that the expression of catechol-O-methyl transferase, an enzyme degrading dopamine, was reduced in the VTA but not in the NAc after FGF21 treatment [84]. These data support the role of FGF21 in modulating dopamine signaling, but more studies are necessary to understand the true relationship between FGF21 and the dopaminergic system.
Interestingly, the FGF21 response to sucrose is associated with body mass index and dorsal striatal signaling in humans, supporting previous evidence in animal models [92].
In conclusion, FGF21 regulates food preference and is proposed to act in areas pretraining the reward system. This fact may open new therapeutic avenues for the use of FGF21 to ameliorate diseases caused by disturbances in homeostatic and hedonic food drives. This assumption is supported by the study of Pena Leon et al. that emphasizes this interconnection by the action of FGF21 on DRd2 in the LHA, an area of the brain that regulates food intake and food reward.

1.5. Therapeutic Use of FGF21

Multiple FGF21 analogues have been developed for treating metabolic diseases [93]. The FGF21 analogue LY2405319 was subcutaneously administered for 28 days to patients with obesity and T2DM, resulting in attenuated dyslipidaemia, reduced body weight and plasma insulin, and increased adiponectin levels [94]. Interestingly, this analogue has also been shown to protect against other derangements induced by the metabolic syndrome, such as neurodegeneration [95]. Moreover, the long-acting FGF21 analogue PF-05231023 caused a marked reduction in serum triglycerides but not in body weight when intravenously administered to hypertriglyceridemic obese people, with or without diabetes, who were already treated with atorvastatin [96]. Further, Pegbelfermin (BMS-986036), a PEGylated human FGF21 analogue, ameliorated dyslipidaemia, increased adiponectin levels, and decreased the levels of fibrosis markers in T2DM patients [97]. Consistently, pegbelfermin considerably reduced the hepatic fat fraction, the markers of hepatic injury, and the biomarkers of fibrosis, and ameliorated dyslipidaemia and increased adiponectin levels, without significantly changing body weight, in a phase IIa clinical trial in NASH patients [98]. Another line of research showed that a bispecific antibody named BFKB8488A that activates the FGFR1/B-klotho complex reduces body weight and induces a sustained improvement in cardiometabolic parameters in obese humans. Interestingly, treatment with BFKB8488A also led to a trend towards a reduction in preference for sweet tastes and carbohydrate intake [99]. Moreover, an analogue of FGF19, another FGF family member (NGM282 or Aldafermin), was tested in NASH patients with promising results [100,101,102,103]. Importantly, in a phase 2 trial, Aldafermin was shown to reduce liver fat and generated a trend towards the improvement of hepatic fibrosis in patients suffering from NASH [103]. Finally, it is worth mentioning that a dual agonist composed by GLP-1 and FGF21 has superior efficacy on body weight control and glycemic control compared to a mixture of GLP-1 and FGF21 [104,105].
Accordingly, in the light of these findings and the success of the unimolecular polypharmacy [106] for the development of a novel generation of drugs (e.g., tirzepatide) to treat obesity and diabetes [107], the development of dual agonists that include FGF21 may provide an interesting tool for the treatment of metabolic diseases.

2. Methods

Study Design, Literature Search and Data Collection

Our aim was to compile the most important literature that provides evidence for how FGF21 regulates metabolism, focusing on the interaction between liver and brain. The review was designed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [108]. Date restrictions were not applied during the literature search. The literature search was primarily conducted using the PubMed database, as well as the reference lists of the selected studies, only for manuscripts written in English. The titles and abstracts of all electronic articles were screened to assess their eligibility.
Our search was performed using the following keywords/MeSH: [“fibroblast growth factor 21” and “obesity”], [“fibroblast growth factor 21” and “feeding behavior”], [“fibroblast growth factor 21” and “liver”], [“fibroblast growth factor 21” and “brain”], [“fibroblast growth factor 21” and “Diabetes Mellitus, Type 2”], [“fibroblast growth factor 21” and “Non-alcoholic Fatty Liver Disease”], [“fibroblast growth factor 21” and “adipose Tissue, Brown”], [“fibroblast growth factor 21” and “Thermogenesis”], [“fibroblast growth factor 21” and “blood glucose”], [“fibroblast growth factor 21” and “insulin resistance”], [“fibroblast growth factor 21” and “humans”]. Two authors (E.P. and S.A.-D) independently screened all articles for eligibility, while potential disagreements were resolved by consensus among all authors.
We sought human studies, experimental studies, and reviews published in medical journals prior to 29 September 2022, while case reports, editorials, conference abstracts, and posters were excluded. Amongst the 7167 published papers considered, 1421 were excluded after duplicate removal and 5746 were excluded during the screening phase because they were out of the review scope as they did not concern FGF21 or not explicitly assess changes in energy balance or glucose homeostasis.
The remaining 750 full-text papers were assessed for eligibility, and 673 were excluded after abstract or full text screening. Finally, 77 studies were selected, plus 27 identified from their reference lists to the review. The flow chart of the selection process is reported in Figure 5.

3. Results and Discussion

The articles referenced in this mini-review focused on general aspects of obesity [1,2,3,4,6,9,10,13,14,21], neuroendocrine control of metabolism [5,7,8,15,16,17,18,19,20,22,23,24,25], non-alcoholic fatty liver disease [11,12], unimolecular polypharmacy [108], and the experimental articles referred to FGF21 expression or referred to the metabolic actions elicited by FGFRs mimetics or activators [26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,107,108].
Among these studies, only 14 were performed on humans, and only eight of these described an FGF analogue or FGF21 signaling activator. In three of them, the compound reduced body weight [94,99,100], while in the other four, no changes in body weight were observed [96,97,98,101]. In addition to the assessment of body weight, four studies found a reduction in peripheral lipid profile [94,96,97,99] and four found a reduction in liver fat content [97,100,101,103] (Table 1).
These variations could be due to the different nature of the compounds, different doses used, routes of administration, or the heterogeneity of the metabolic state of the patients. All together, we can conclude that FGF21 analogues display an a priori high applicability; however, although compounds based on FGF21 seem to be promising targets against metabolic diseases such as T2DM and NASH, their effect on body weight is still suboptimal.
The quality assessment was performed by the Cochrane risk-of-bias tool for randomized trial RoB scale in the manuscript regarding only clinical trials (Supplemental Table S1), while the outcomes of the qualitative synthesis are presented in Table 2.

4. Concluding Remarks

In this review, we covered the metabolic actions of FGF21 and its effect on energy balance. This hepatic signal appears not only as a key metabolic factor directly implicated in T2DM and non-alcoholic fatty liver disease (NAFLD)/NASH by its action in glucose and lipid metabolism but also as regulators of food intake and body weight through its action on the CNS. Regardless of the great expectations for the use of FGF21 as a therapeutic target, there are differences in the physiological functions of FGF21 between mice and humans, a fact that complicates the translational value of the peptide [109].
Therefore, in spite of the recent advances in defining the intracellular function and regulation of FGF21, the molecular mechanisms mediating its actions and its physiological and pathophysiological roles in CNS homeostasis are not totally understood.
In conclusion, FGF21 continues to emerge as an interesting tool for the study and precise understanding of metabolic and liver diseases.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/ijms232113318/s1.

Author Contributions

M.Q. and O.A.-M., conceived the manuscript; E.P., S.Á.-D., R.P.-L., M.S.-T., A.E.-G., J.F., L.M.S., M.Q. and O.A.-M., wrote the manuscript. All authors made substantial contributions to the discussion of the content and reviewed/edited the manuscript before submission. All authors have read and agreed to the published version of the manuscript. All authors drafting the work or revising it critically for important intellectual content before submission.

Funding

This study has been funded by Instituto de Salud Carlos III (ISCIII) through the grant number PI18/00998 (L.M.S.) and PI21/01216 (O.A.-M.) and co-funded by the European Union. GAIN Proyectos de Excelencia with the grants numbers IN607D-2022-07 (O.A.-M.) and IN607D-2022-04 (L.M.S.). Fundación de la Sociedad Gallega de Endocrinología y Nutrición (O.A.-M.); Centro de Investigación Biomédica en Red (CIBER) de Fisiopatología de la Obesidad y Nutrición (CIBERobn). CIBERobn is an initiative of the Instituto de Salud Carlos III (ISCIII) of Spain, which is supported by FEDER funds; O.A.-M. and M.Q. were funded by a research contract Miguel Servet (CP20/00146 and CP21/00108, respectively) from the ISCIII co-funded by the European Union. The data figures were generated by using materials from Servier Medical Art (Servier) under consideration of a Creative Commons Attribution 3.0 Unported License.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Farooqi, S.; O’Rahilly, S. Genetics of Obesity in Humans. Endocr. Rev. 2006, 27, 710–718. [Google Scholar] [CrossRef] [PubMed]
  2. Jéquier, E. Pathways to obesity. Int. J. Obes. 2002, 26, S12–S17. [Google Scholar] [CrossRef] [Green Version]
  3. NCD Risk Factor Collaboration (NCD-RisC) Rising rural body-mass index is the main driver of the global obesity epidemic in adults. Nature 2019, 569, 260–264. [CrossRef] [PubMed] [Green Version]
  4. Hill, J.O.; Peters, J.C. Environmental Contributions to the Obesity Epidemic. Science 1998, 280, 1371–1374. [Google Scholar] [CrossRef] [PubMed]
  5. Berthoud, H.-R.; Morrison, C. The Brain, Appetite, and Obesity. Annu. Rev. Psychol. 2008, 59, 55–92. [Google Scholar] [CrossRef] [PubMed]
  6. Foster, G.D.; Wadden, T.A.; Kendall, P.C.; Stunkard, A.J.; Al, E. Psychological effects of weight loss and regain: A prospective evaluation. J. Consult. Clin. Psychol. 1996, 64, 752–757. [Google Scholar] [CrossRef]
  7. Broberger, C. Brain regulation of food intake and appetite: Molecules and networks. J. Intern. Med. 2005, 258, 301–327. [Google Scholar] [CrossRef]
  8. Jais, A.; Brüning, J.C. Arcuate Nucleus-Dependent Regulation of Metabolism—Pathways to Obesity and Diabetes Mellitus. Endocr. Rev. 2021, 43, 314–328. [Google Scholar] [CrossRef]
  9. Grundy, S. Metabolic Syndrome: Therapeutic Considerations. Handb. Exp. Pharmacol. 2005, 170, 107–133. [Google Scholar] [CrossRef]
  10. Huang, P.L. A comprehensive definition for metabolic syndrome. Dis. Model. Mech. 2009, 2, 231–237. [Google Scholar] [CrossRef]
  11. Angulo, P. Nonalcoholic Fatty Liver Disease. N. Engl. J. Med. 2002, 346, 1221–1231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Quiñones, M.; Fernø, J.; Al-Massadi, O. Ghrelin and liver disease. Rev. Endocr. Metab. Disord. 2019, 21, 45–56. [Google Scholar] [CrossRef] [PubMed]
  13. Quiñones, M.; Fernø, J.; Diéguez, C.; Nogueiras, R.; Al-Massadi, O. Exciting advances in GPCR-based drugs discovery for treating metabolic disease and future perspectives. Expert Opin. Drug Discov. 2019, 14, 421–431. [Google Scholar] [CrossRef] [PubMed]
  14. Al Massadi, O.; Nogueiras, R.; Dieguez, C.; Girault, J.-A. Ghrelin and food reward. Neuropharmacology 2019, 148, 131–138. [Google Scholar] [CrossRef]
  15. Prida, E.; Fernández-González, S.; Pena-León, V.; Pérez-Lois, R.; Fernø, J.; Seoane, L.M.; Quiñones, M.; Al Massadi, O. Crosstalk between Melanin Concentrating Hormone and Endocrine Factors: Implications for Obesity. Int. J. Mol. Sci. 2022, 23, 2436. [Google Scholar] [CrossRef]
  16. Saper, C.B.; Chou, T.C.; Elmquist, J.K. The Need to Feed: Homeostatic and Hedonic Control of Eating. Neuron 2002, 36, 199–211. [Google Scholar] [CrossRef] [Green Version]
  17. Schneeberger, M.; Gomis, R.; Claret, M. Hypothalamic and brainstem neuronal circuits controlling homeostatic energy balance. J. Endocrinol. 2014, 220, T25–T46. [Google Scholar] [CrossRef] [Green Version]
  18. Al-Massadi, O.; Dieguez, C.; Schneeberger, M.; López, M.; Schwaninger, M.; Prevot, V.; Nogueiras, R. Multifaceted actions of melanin-concentrating hormone on mammalian energy homeostasis. Nat. Rev. Endocrinol. 2021, 17, 745–755. [Google Scholar] [CrossRef]
  19. Al Massadi, O.; López, M.; Tschöp, M.; Diéguez, C.; Nogueiras, R. Current Understanding of the Hypothalamic Ghrelin Pathways Inducing Appetite and Adiposity. Trends Neurosci. 2017, 40, 167–180. [Google Scholar] [CrossRef]
  20. Timper, K.; Brüning, J.C. Hypothalamic circuits regulating appetite and energy homeostasis: Pathways to obesity. Dis. Model. Mech. 2017, 10, 679–689. [Google Scholar] [CrossRef]
  21. Friedman, J.M. Modern science versus the stigma of obesity. Nat. Med. 2004, 10, 563–569. [Google Scholar] [CrossRef] [PubMed]
  22. Myers, M.G.; Olson, D. Central nervous system control of metabolism. Nature 2012, 491, 357–363. [Google Scholar] [CrossRef] [PubMed]
  23. Valassi, E.; Scacchi, M.; Cavagnini, F. Neuroendocrine control of food intake. Nutr. Metab. Cardiovasc. Dis. 2008, 18, 158–168. [Google Scholar] [CrossRef]
  24. Quarta, C.; Claret, M.; Zeltser, L.M.; Williams, K.W.; Yeo, G.S.H.; Tschöp, M.H.; Diano, S.; Brüning, J.C.; Cota, D. POMC neuronal heterogeneity in energy balance and beyond: An integrated view. Nat. Metab. 2021, 3, 299–308. [Google Scholar] [CrossRef] [PubMed]
  25. Yeo, G.S.; Chao, D.H.M.; Siegert, A.-M.; Koerperich, Z.M.; Ericson, M.D.; Simonds, S.E.; Larson, C.M.; Luquet, S.; Clarke, I.; Sharma, S.; et al. The melanocortin pathway and energy homeostasis: From discovery to obesity therapy. Mol. Metab. 2021, 48, 101206. [Google Scholar] [CrossRef] [PubMed]
  26. Nishimura, T.; Nakatake, Y.; Konishi, M.; Itoh, N. Identification of a novel FGF, FGF-21, preferentially expressed in the liver. Biochim. Et Biophys. Acta (BBA)-Gene Struct. Expr. 2000, 1492, 203–206. [Google Scholar] [CrossRef]
  27. Johnson, C.L.; Weston, J.Y.; Chadi, S.A.; Fazio, E.N.; Huff, M.W.; Kharitonenkov, A.; Köester, A.; Pin, C.L. Fibroblast Growth Factor 21 Reduces the Severity of Cerulein-Induced Pancreatitis in Mice. Gastroenterology 2009, 137, 1795–1804. [Google Scholar] [CrossRef]
  28. Markan, K.R.; Naber, M.C.; Ameka, M.K.; Anderegg, M.D.; Mangelsdorf, D.J.; Kliewer, S.A.; Mohammadi, M.; Potthoff, M.J. Circulating FGF21 Is Liver Derived and Enhances Glucose Uptake During Refeeding and Overfeeding. Diabetes 2014, 63, 4057–4063. [Google Scholar] [CrossRef] [Green Version]
  29. Fisher, F.M.; Maratos-Flier, E. Understanding the Physiology of FGF21. Annu. Rev. Physiol. 2016, 78, 223–241. [Google Scholar] [CrossRef] [Green Version]
  30. Fisher, F.M.; Kleiner, S.; Douris, N.; Fox, E.C.; Mepani, R.J.; Verdeguer, F.; Wu, J.; Kharitonenkov, A.; Flier, J.S.; Maratos-Flier, E.; et al. FGF21 regulates PGC-1α and browning of white adipose tissues in adaptive thermogenesis. Genes Dev. 2012, 26, 271–281. [Google Scholar] [CrossRef]
  31. Coate, K.C.; Hernandez, G.; Thorne, C.A.; Sun, S.; Le, T.D.; Vale, K.; Kliewer, S.A.; Mangelsdorf, D.J. FGF21 Is an Exocrine Pancreas Secretagogue. Cell Metab. 2017, 25, 472–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Inagaki, T.; Dutchak, P.; Zhao, G.; Ding, X.; Gautron, L.; Parameswara, V.; Li, Y.; Goetz, R.; Mohammadi, M.; Esser, V.; et al. Endocrine Regulation of the Fasting Response by PPARα-Mediated Induction of Fibroblast Growth Factor 21. Cell Metab. 2007, 5, 415–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Badman, M.K.; Pissios, P.; Kennedy, A.R.; Koukos, G.; Flier, J.S.; Maratos-Flier, E. Hepatic Fibroblast Growth Factor 21 Is Regulated by PPARα and Is a Key Mediator of Hepatic Lipid Metabolism in Ketotic States. Cell Metab. 2007, 5, 426–437. [Google Scholar] [CrossRef] [Green Version]
  34. Lin, X.; Liu, Y.B.; Hu, H. Metabolic role of fibroblast growth factor 21 in liver, adipose and nervous system tissues. Biomed. Rep. 2017, 6, 495–502. [Google Scholar] [CrossRef] [Green Version]
  35. Ding, X.; Boney-Montoya, J.; Owen, B.; Bookout, A.L.; Coate, K.; Mangelsdorf, D.J.; Kliewer, S.A. βKlotho Is Required for Fibroblast Growth Factor 21 Effects on Growth and Metabolism. Cell Metab. 2012, 16, 387–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Tacer, K.F.; Bookout, A.L.; Ding, X.; Kurosu, H.; John, G.B.; Wang, L.; Goetz, R.; Mohammadi, M.; Kuro-O, M.; Mangelsdorf, D.J.; et al. Research Resource: Comprehensive Expression Atlas of the Fibroblast Growth Factor System in Adult Mouse. Mol. Endocrinol. 2010, 24, 2050–2064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Bookout, A.L.; De Groot, M.H.M.; Owen, B.; Lee, S.; Gautron, L.; Lawrence, H.L.; Ding, X.; Elmquist, J.K.; Takahashi, J.; Mangelsdorf, D.; et al. FGF21 regulates metabolism and circadian behavior by acting on the nervous system. Nat. Med. 2013, 19, 1147–1152. [Google Scholar] [CrossRef]
  38. Kurosu, H.; Choi, M.; Ogawa, Y.; Dickson, A.S.; Goetz, R.; Eliseenkova, A.V.; Mohammadi, M.; Rosenblatt, K.P.; Kliewer, S.A.; Kuro-O, M. Tissue-specific Expression of βKlotho and Fibroblast Growth Factor (FGF) Receptor Isoforms Determines Metabolic Activity of FGF19 and FGF21. J. Biol. Chem. 2007, 282, 26687–26695. [Google Scholar] [CrossRef] [Green Version]
  39. Ogawa, Y.; Kurosu, H.; Yamamoto, M.; Nandi, A.; Rosenblatt, K.P.; Goetz, R.; Eliseenkova, A.V.; Mohammadi, M.; Kuro-O, M. βKlotho is required for metabolic activity of fibroblast growth factor 21. Proc. Natl. Acad. Sci. USA 2007, 104, 7432–7437. [Google Scholar] [CrossRef] [Green Version]
  40. Suzuki, M.; Uehara, Y.; Motomura-Matsuzaka, K.; Oki, J.; Koyama, Y.; Kimura, M.; Asada, M.; Komi-Kuramochi, A.; Oka, S.; Toru, T. βKlotho Is Required for Fibroblast Growth Factor (FGF) 21 Signaling through FGF Receptor (FGFR) 1c and FGFR3c. Mol. Endocrinol. 2008, 22, 1006–1014. [Google Scholar] [CrossRef]
  41. Bono, B.S.; Ly, N.K.K.; Miller, P.A.; Williams-Ikhenoba, J.; Dumiaty, Y.; Chee, M.J. Spatial distribution of beta-klotho mRNA in the mouse hypothalamus, hippocampal region, subiculum, and amygdala. J. Comp. Neurol. 2022, 530, 1634–1657. [Google Scholar] [CrossRef] [PubMed]
  42. Hultman, K.; Scarlett, J.M.; Baquero, A.F.; Cornea, A.; Zhang, Y.; Salinas, C.B.G.; Brown, J.; Morton, G.J.; Whalen, E.J.; Grove, K.L.; et al. The central fibroblast growth factor receptor/beta klotho system: Comprehensive mapping in Mus musculus and comparisons to nonhuman primate and human samples using an automated in situ hybridization platform. J. Comp. Neurol. 2019, 527, 2069–2085. [Google Scholar] [CrossRef] [PubMed]
  43. Potthoff, M.J.; Inagaki, T.; Satapati, S.; Ding, X.; He, T.; Goetz, R.; Mohammadi, M.; Finck, B.N.; Mangelsdorf, D.J.; Kliewer, S.A.; et al. FGF21 induces PGC-1α and regulates carbohydrate and fatty acid metabolism during the adaptive starvation response. Proc. Natl. Acad. Sci. USA 2009, 106, 10853–10858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Kim, E.R.; Kim, S.R.; Cho, W.; Lee, S.-G.; Kim, S.H.; Kim, J.H.; Choi, E.; Kim, J.-H.; Yu, J.-W.; Lee, B.-W.; et al. Short Term Isocaloric Ketogenic Diet Modulates NLRP3 Inflammasome Via B-hydroxybutyrate and Fibroblast Growth Factor 21. Front. Immunol. 2022, 13, 845352. [Google Scholar] [CrossRef] [PubMed]
  45. Forney, L.A.; Fang, H.; Sims, L.C.; Stone, K.P.; Vincik, L.Y.; Vick, A.M.; Gibson, A.N.; Burk, D.H.; Gettys, T.W. Dietary Methionine Restriction Signals to the Brain Through Fibroblast Growth Factor 21 to Regulate Energy Balance and Remodeling of Adipose Tissue. Obesity 2020, 28, 1912–1921. [Google Scholar] [CrossRef]
  46. Moro, J.; Chaumontet, C.; Even, P.C.; Blais, A.; Piedcoq, J.; Gaudichon, C.; Tomé, D.; Azzout-Marniche, D. Severe protein deficiency induces hepatic expression and systemic level of FGF21 but inhibits its hypothalamic expression in growing rats. Sci. Rep. 2021, 11, 12436. [Google Scholar] [CrossRef]
  47. Zhang, X.; Yeung, D.C.Y.; Karpisek, M.; Stejskal, D.; Zhou, Z.-G.; Liu, F.; Wong, R.L.C.; Chow, W.-S.; Tso, A.W.K.; Lam, K.S.L.; et al. Serum FGF21 Levels Are Increased in Obesity and Are Independently Associated with the Metabolic Syndrome in Humans. Diabetes 2008, 57, 1246–1253. [Google Scholar] [CrossRef] [Green Version]
  48. Mraz, M.; Bartlova, M.; Lacinova, Z.; Michalsky, D.; Kasalicky, M.; Haluzikova, D.; Matoulek, M.; Dostalova, I.; Humenanska, V.; Haluzik, M. Serum concentrations and tissue expression of a novel endocrine regulator fibroblast growth factor-21 in patients with type 2 diabetes and obesity. Clin. Endocrinol. 2009, 71, 369–375. [Google Scholar] [CrossRef]
  49. Dushay, J.; Chui, P.C.; Gopalakrishnan, G.S.; Varela–Rey, M.; Crawley, M.; Fisher, F.M.; Badman, M.K.; Chantar, M.L.M.; Maratos–Flier, E. Increased Fibroblast Growth Factor 21 in Obesity and Nonalcoholic Fatty Liver Disease. Gastroenterology 2010, 139, 456–463. [Google Scholar] [CrossRef] [Green Version]
  50. Kiluk, P.; Baran, A.; Kaminski, T.W.; Maciaszek, M.; Flisiak, I. The Level of FGF 21 as a New Risk Factor for the Occurrence of Cardiometabolic Disorders amongst the Psoriatic Patients. J. Clin. Med. 2019, 8, 2206. [Google Scholar] [CrossRef]
  51. Miehle, K.; Ebert, T.; Kralisch, S.; Hoffmann, A.; Kratzsch, J.; Schlögl, H.; Stumvoll, M.; Fasshauer, M. Serum concentrations of fibroblast growth factor 21 are elevated in patients with congenital or acquired lipodystrophy. Cytokine 2016, 83, 239–244. [Google Scholar] [CrossRef] [PubMed]
  52. Lindegaard, B.; Hvid, T.; Grøndahl, T.; Frosig, C.; Gerstoft, J.; Hojman, P.; Pedersen, B.K. Expression of Fibroblast Growth Factor-21 in Muscle Is Associated with Lipodystrophy, Insulin Resistance and Lipid Disturbances in Patients with HIV. PLoS ONE 2013, 8, e55632. [Google Scholar] [CrossRef]
  53. Rodríguez-Nuevo, A.; Díaz-Ramos, A.; Noguera, E.; Díaz-Sáez, F.; Duran, X.; Muñoz, J.P.; Romero, M.; Plana, N.; Sebastián, D.; Tezze, C.; et al. Mitochondrial DNA and TLR9 drive muscle inflammation upon Opa1 deficiency. EMBO J. 2018, 37, e96553. [Google Scholar] [CrossRef]
  54. Coskun, T.; Bina, H.A.; Schneider, M.A.; Dunbar, J.D.; Hu, C.C.; Chen, Y.; Moller, D.E.; Kharitonenkov, A. Fibroblast Growth Factor 21 Corrects Obesity in Mice. Endocrinology 2008, 149, 6018–6027. [Google Scholar] [CrossRef]
  55. Kharitonenkov, A.; Shiyanova, T.L.; Koester, A.; Ford, A.M.; Micanovic, R.; Galbreath, E.J.; Sandusky, G.E.; Hammond, L.J.; Moyers, J.S.; Owens, R.A.; et al. FGF-21 as a novel metabolic regulator. J. Clin. Investig. 2005, 115, 1627–1635. [Google Scholar] [CrossRef] [Green Version]
  56. Tomlinson, E.; Fu, L.; John, L.; Hultgren, B.; Huang, X.; Renz, M.; Stephan, J.P.; Tsai, S.P.; Powell-Braxton, L.; French, D.; et al. Transgenic Mice Expressing Human Fibroblast Growth Factor-19 Display Increased Metabolic Rate and Decreased Adiposity. Endocrinology 2002, 143, 1741–1747. [Google Scholar] [CrossRef]
  57. Xu, J.; Stanislaus, S.; Chinookoswong, N.; Lau, Y.Y.; Hager, T.; Patel, J.; Ge, H.; Weiszmann, J.; Lu, S.-C.; Graham, M.; et al. Acute glucose-lowering and insulin-sensitizing action of FGF21 in insulin-resistant mouse models—Association with liver and adipose tissue effects. Am. J. Physiol. Metab. 2009, 297, E1105–E1114. [Google Scholar] [CrossRef] [Green Version]
  58. Kharitonenkov, A.; Wroblewski, V.J.; Koester, A.; Chen, Y.-F.; Clutinger, C.K.; Tigno, X.T.; Hansen, B.C.; Shanafelt, A.B.; Etgen, G.J. The Metabolic State of Diabetic Monkeys Is Regulated by Fibroblast Growth Factor-21. Endocrinology 2007, 148, 774–781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Adams, A.C.; Halstead, C.A.; Hansen, B.C.; Irizarry, A.R.; Martin, J.A.; Myers, S.R.; Reynolds, V.L.; Smith, H.W.; Wroblewski, V.J.; Kharitonenkov, A. LY2405319, an Engineered FGF21 Variant, Improves the Metabolic Status of Diabetic Monkeys. PLoS ONE 2013, 8, e65763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Andersen, B.; Straarup, E.M.; Heppner, K.M.; Takahashi, D.L.; Raffaele, V.; Dissen, G.A.; Lewandowski, K.; Bödvarsdottir, T.B.; Raun, K.; Grove, K.L.; et al. FGF21 decreases body weight without reducing food intake or bone mineral density in high-fat fed obese rhesus macaque monkeys. Int. J. Obes. 2018, 42, 1151–1160. [Google Scholar] [CrossRef]
  61. Dutchak, P.A.; Katafuchi, T.; Bookout, A.L.; Choi, J.H.; Yu, R.T.; Mangelsdorf, D.J.; Kliewer, S.A. Fibroblast Growth Factor-21 Regulates PPARγ Activity and the Antidiabetic Actions of Thiazolidinediones. Cell 2012, 148, 556–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Li, H.; Gao, Z.; Zhang, J.; Ye, X.; Xu, A.; Ye, J.; Jia, W. Sodium Butyrate Stimulates Expression of Fibroblast Growth Factor 21 in Liver by Inhibition of Histone Deacetylase 3. Diabetes 2012, 61, 797–806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Veniant, M.M.; Hale, C.; Helmering, J.; Chen, M.M.; Stanislaus, S.; Busby, J.; Vonderfecht, S.; Xu, J.; Lloyd, D.J. FGF21 Promotes Metabolic Homeostasis via White Adipose and Leptin in Mice. PLoS ONE 2012, 7, e40164. [Google Scholar] [CrossRef]
  64. Adams, A.C.; Coskun, T.; Cheng, C.C.; O′farrell, L.S.; DuBois, S.L.; Kharitonenkov, A. Fibroblast growth factor 21 is not required for the antidiabetic actions of the thiazoladinediones. Mol. Metab. 2013, 2, 205–214. [Google Scholar] [CrossRef] [PubMed]
  65. Lin, Z.; Tian, H.; Lam, K.S.; Lin, S.; Hoo, R.C.; Konishi, M.; Itoh, N.; Wang, Y.; Bornstein, S.R.; Xu, A.; et al. Adiponectin Mediates the Metabolic Effects of FGF21 on Glucose Homeostasis and Insulin Sensitivity in Mice. Cell Metab. 2013, 17, 779–789. [Google Scholar] [CrossRef] [Green Version]
  66. Holland, W.L.; Adams, A.C.; Brozinick, J.T.; Bui, H.H.; Miyauchi, Y.; Kusminski, C.M.; Bauer, S.M.; Wade, M.; Singhal, E.; Cheng, C.C.; et al. An FGF21-Adiponectin-Ceramide Axis Controls Energy Expenditure and Insulin Action in Mice. Cell Metab. 2013, 17, 790–797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Hondares, E.; Rosell, M.; Gonzalez, F.J.; Giralt, M.; Iglesias, R.; Villarroya, F. Hepatic FGF21 Expression Is Induced at Birth via PPARα in Response to Milk Intake and Contributes to Thermogenic Activation of Neonatal Brown Fat. Cell Metab. 2010, 11, 206–212. [Google Scholar] [CrossRef] [Green Version]
  68. Chen, W.; Hoo, R.L.-C.; Konishi, M.; Itoh, N.; Lee, P.-C.; Ye, H.-Y.; Lam, K.S.-L.; Xu, A. Growth Hormone Induces Hepatic Production of Fibroblast Growth Factor 21 through a Mechanism Dependent on Lipolysis in Adipocytes. J. Biol. Chem. 2011, 286, 34559–34566. [Google Scholar] [CrossRef] [Green Version]
  69. Hondares, E.; Iglesias, R.; Giralt, A.; Gonzalez, F.J.; Giralt, M.; Mampel, T.; Villarroya, F. Thermogenic Activation Induces FGF21 Expression and Release in Brown Adipose Tissue. J. Biol. Chem. 2011, 286, 12983–12990. [Google Scholar] [CrossRef] [Green Version]
  70. Potthoff, M.J.; Kliewer, S.A.; Mangelsdorf, D.J. Endocrine fibroblast growth factors 15/19 and 21: From feast to famine. Genes Dev. 2012, 26, 312–324. [Google Scholar] [CrossRef]
  71. Douris, N.; Stevanovic, D.M.; Fisher, F.M.; Cisu, T.I.; Chee, M.; Nguyen, N.L.; Zarebidaki, E.; Adams, A.C.; Kharitonenkov, A.; Flier, J.S.; et al. Central Fibroblast Growth Factor 21 Browns White Fat via Sympathetic Action in Male Mice. Endocrinology 2015, 156, 2470–2481. [Google Scholar] [CrossRef] [PubMed]
  72. Owen, B.M.; Ding, X.; Morgan, D.A.; Coate, K.C.; Bookout, A.L.; Rahmouni, K.; Kliewer, S.A.; Mangelsdorf, D.J. FGF21 Acts Centrally to Induce Sympathetic Nerve Activity, Energy Expenditure, and Weight Loss. Cell Metab. 2014, 20, 670–677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Lan, T.; Morgan, D.A.; Rahmouni, K.; Sonoda, J.; Fu, X.; Burgess, S.C.; Holland, W.L.; Kliewer, S.A.; Mangelsdorf, D.J. FGF19, FGF21, and an FGFR1/β-Klotho-Activating Antibody Act on the Nervous System to Regulate Body Weight and Glycemia. Cell Metab. 2017, 26, 709–718.e3. [Google Scholar] [CrossRef]
  74. Marcelin, G.; Jo, Y.-H.; Li, X.; Schwartz, G.J.; Zhang, Y.; Dun, N.J.; Lyu, R.-M.; Blouet, C.; Chang, J.K.; Chua, S., Jr. Central action of FGF19 reduces hypothalamic AGRP/NPY neuron activity and improves glucose metabolism. Mol. Metab. 2014, 3, 19–28. [Google Scholar] [CrossRef] [PubMed]
  75. Mutsnaini, L.; Kim, C.-S.; Kim, J.; Joe, Y.; Chung, H.T.; Choi, H.-S.; Roh, E.; Kim, M.-S.; Yu, R. Fibroblast growth factor 21 deficiency aggravates obesity-induced hypothalamic inflammation and impairs thermogenic response. Agents Actions 2019, 68, 351–358. [Google Scholar] [CrossRef]
  76. Liang, Q.; Zhong, L.; Zhang, J.; Wang, Y.; Bornstein, S.R.; Triggle, C.R.; Ding, H.; Lam, K.S.; Xu, A. FGF21 Maintains Glucose Homeostasis by Mediating the Cross Talk Between Liver and Brain During Prolonged Fasting. Diabetes 2014, 63, 4064–4075. [Google Scholar] [CrossRef] [Green Version]
  77. Inagaki, T.; Lin, V.Y.; Goetz, R.; Mohammadi, M.; Mangelsdorf, D.; Kliewer, S.A. Inhibition of Growth Hormone Signaling by the Fasting-Induced Hormone FGF21. Cell Metab. 2008, 8, 77–83. [Google Scholar] [CrossRef] [Green Version]
  78. Hill, C.M.; Laeger, T.; Dehner, M.; Albarado, D.C.; Clarke, B.; Wanders, D.; Burke, S.J.; Collier, J.J.; Qualls-Creekmore, E.; Solon-Biet, S.M.; et al. FGF21 Signals Protein Status to the Brain and Adaptively Regulates Food Choice and Metabolism. Cell Rep. 2019, 27, 2934–2947.e3. [Google Scholar] [CrossRef] [Green Version]
  79. Blanco, A.M.; Bertucci, J.I.; Unniappan, S. FGF21 Mimics a Fasting-Induced Metabolic State and Increases Appetite in Zebrafish. Sci. Rep. 2020, 10, 6993. [Google Scholar] [CrossRef] [Green Version]
  80. Song, P.; Zechner, C.; Hernandez, G.; Cánovas, J.; Xie, Y.; Sondhi, V.; Wagner, M.; Stadlbauer, V.; Horvath, A.; Leber, B.; et al. The Hormone FGF21 Stimulates Water Drinking in Response to Ketogenic Diet and Alcohol. Cell Metab. 2018, 27, 1338–1347.e4. [Google Scholar] [CrossRef]
  81. Hsuchou, H.; Pan, W.; Kastin, A.J. The fasting polypeptide FGF21 can enter brain from blood. Peptides 2007, 28, 2382–2386. [Google Scholar] [CrossRef] [Green Version]
  82. von Holstein-Rathlou, S.; BonDurant, L.D.; Peltekian, L.; Naber, M.C.; Yin, T.C.; Claflin, K.E.; Urizar, A.I.; Madsen, A.N.; Ratner, C.; Holst, B.; et al. FGF21 Mediates Endocrine Control of Simple Sugar Intake and Sweet Taste Preference by the Liver. Cell Metab. 2015, 23, 335–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Larson, K.R.; Chaffin, A.T.-B.; Goodson, M.L.; Fang, Y.; Ryan, K.K. Fibroblast Growth Factor-21 Controls Dietary Protein Intake in Male Mice. Endocrinology 2019, 160, 1069–1080. [Google Scholar] [CrossRef] [PubMed]
  84. Talukdar, S.; Owen, B.M.; Song, P.; Hernandez, G.; Zhang, Y.; Zhou, Y.; Scott, W.T.; Paratala, B.; Turner, T.; Smith, A.; et al. FGF21 Regulates Sweet and Alcohol Preference. Cell Metab. 2015, 23, 344–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Jensen-Cody, S.O.; Flippo, K.H.; Claflin, K.E.; Yavuz, Y.; Sapouckey, S.A.; Walters, G.C.; Usachev, Y.M.; Atasoy, D.; Gillum, M.P.; Potthoff, M.J. FGF21 Signals to Glutamatergic Neurons in the Ventromedial Hypothalamus to Suppress Carbohydrate Intake. Cell Metab. 2020, 32, 273–286.e6. [Google Scholar] [CrossRef] [PubMed]
  86. Flippo, K.H.; Jensen-Cody, S.O.; Claflin, K.E.; Potthoff, M.J. FGF21 signaling in glutamatergic neurons is required for weight loss associated with dietary protein dilution. Sci. Rep. 2020, 10, 19521. [Google Scholar] [CrossRef] [PubMed]
  87. Recinella, L.; Leone, S.; Ferrante, C.; Chiavaroli, A.; Di Nisio, C.; Martinotti, S.; Vacca, M.; Brunetti, L.; Orlando, G. Effects of central fibroblast growth factor 21 (FGF21) in energy balance. J. Biol. Regul. Homeost. Agents 2017, 31, 603–613. [Google Scholar]
  88. Špolcová, A.; Holubová, M.; Mikulášková, B.; Nagelová, V.; Štofková, A.; Lacinová, Z.; Jurčovičová, J.; Haluzik, M.; Maletínská, L.; Železná, B. Changes in FGF21 Serum Concentrations and Liver mRNA Expression in an Experimental Model of Complete Lipodystrophy and Insulin-Resistant Diabetes. Physiol. Res. 2014, 63, 483–490. [Google Scholar] [CrossRef]
  89. Geller, S.; Arribat, Y.; Netzahualcoyotzi, C.; Lagarrigue, S.; Carneiro, L.; Zhang, L.; Amati, F.; Lopez-Mejia, I.C.; Pellerin, L. Tanycytes Regulate Lipid Homeostasis by Sensing Free Fatty Acids and Signaling to Key Hypothalamic Neuronal Populations via FGF21 Secretion. Cell Metab. 2019, 30, 833–844.e7. [Google Scholar] [CrossRef]
  90. Pena-Leon, V.; Folgueira, C.; Barja-Fernández, S.; Pérez-Lois, R.; Lima, N.D.S.; Martin, M.; Heras, V.; Martinez-Martinez, S.; Valero, P.; Iglesias, C.; et al. Prolonged breastfeeding protects from obesity by hypothalamic action of hepatic FGF21. Nat. Metab. 2022, 4, 901–917. [Google Scholar] [CrossRef]
  91. Zhou, B.; Claflin, K.E.; Flippo, K.H.; Sullivan, A.I.; Asghari, A.; Tadinada, S.M.; Jensen-Cody, S.O.; Abel, T.; Potthoff, M.J. Central FGF21 production regulates memory but not peripheral metabolism. Cell Rep. 2022, 40, 111239. [Google Scholar] [CrossRef] [PubMed]
  92. Alves, J.M.; Yunker, A.G.; Luo, S.; Jann, K.; Angelo, B.; DeFendis, A.; Pickering, T.A.; Smith, A.; Monterosso, J.R.; Page, K.A. FGF21 response to sucrose is associated with BMI and dorsal striatal signaling in humans. Obesity 2022, 30, 1239–1247. [Google Scholar] [CrossRef] [PubMed]
  93. Gimeno, R.E.; Moller, D.E. FGF21-based pharmacotherapy—Potential utility for metabolic disorders. Trends Endocrinol. Metab. 2014, 25, 303–311. [Google Scholar] [CrossRef] [PubMed]
  94. Gaich, G.; Chien, J.Y.; Fu, H.; Glass, L.C.; Deeg, M.A.; Holland, W.L.; Kharitonenkov, A.; Bumol, T.; Schilske, H.K.; Moller, D.E. The Effects of LY2405319, an FGF21 Analog, in Obese Human Subjects with Type 2 Diabetes. Cell Metab. 2013, 18, 333–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Rühlmann, C.; Dannehl, D.; Brodtrück, M.; Adams, A.C.; Stenzel, J.; Lindner, T.; Krause, B.J.; Vollmar, B.; Kuhla, A. Neuroprotective Effects of the FGF21 Analogue LY2405319. J. Alzheimer’s Dis. 2021, 80, 357–369. [Google Scholar] [CrossRef]
  96. Kim, A.; Somayaji, V.R.; Dong, J.Q.; Rolph, T.P.; Weng, Y.; Chabot, J.R.; Gropp, K.E.; Talukdar, S.; Calle, R.A. Once-weekly administration of a long-acting fibroblast growth factor 21 analogue modulates lipids, bone turnover markers, blood pressure and body weight differently in obese people with hypertriglyceridaemia and in non-human primates. Diabetes Obes. Metab. 2017, 19, 1762–1772. [Google Scholar] [CrossRef]
  97. Charles, E.D.; Neuschwander-Tetri, B.A.; Frias, J.P.; Kundu, S.; Luo, Y.; Tirucherai, G.S.; Christian, R. Pegbelfermin (BMS-986036), PEGylated FGF21, in Patients with Obesity and Type 2 Diabetes: Results from a Randomized Phase 2 Study. Obesity 2018, 27, 41–49. [Google Scholar] [CrossRef] [Green Version]
  98. Sanyal, A.; Charles, E.D.; A Neuschwander-Tetri, B.; Loomba, R.; A Harrison, S.; Abdelmalek, M.F.; Lawitz, E.J.; Halegoua-DeMarzio, D.; Kundu, S.; Noviello, S.; et al. Pegbelfermin (BMS-986036), a PEGylated fibroblast growth factor 21 analogue, in patients with non-alcoholic steatohepatitis: A randomised, double-blind, placebo-controlled, phase 2a trial. Lancet 2018, 392, 2705–2717. [Google Scholar] [CrossRef]
  99. Baruch, A.; Wong, C.; Chinn, L.W.; Vaze, A.; Sonoda, J.; Gelzleichter, T.; Chen, S.; Lewin-Koh, N.; Morrow, L.; Dheerendra, S.; et al. Antibody-mediated activation of the FGFR1/Klothoβ complex corrects metabolic dysfunction and alters food preference in obese humans. Proc. Natl. Acad. Sci. USA 2020, 117, 28992–29000. [Google Scholar] [CrossRef]
  100. Harrison, S.A.; Rinella, M.E.; Abdelmalek, M.F.; Trotter, J.F.; Paredes, A.H.; Arnold, H.L.; Kugelmas, M.; Bashir, M.R.; Jaros, M.J.; Ling, L.; et al. NGM282 for treatment of non-alcoholic steatohepatitis: A multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2018, 391, 1174–1185. [Google Scholar] [CrossRef]
  101. Harrison, S.A.; Rossi, S.J.; Paredes, A.H.; Trotter, J.F.; Bashir, M.R.; Guy, C.D.; Banerjee, R.; Jaros, M.J.; Owers, S.; Baxter, B.A.; et al. NGM282 Improves Liver Fibrosis and Histology in 12 Weeks in Patients with Nonalcoholic Steatohepatitis. Hepatology 2019, 71, 1198–1212. [Google Scholar] [CrossRef]
  102. Tabibian, J.H.; Lindor, K.D. NGM282, an FGF19 analogue, in primary sclerosing cholangitis: A nebulous matter. J. Hepatol. 2019, 70, 348–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Harrison, S.A.; Neff, G.; Guy, C.D.; Bashir, M.R.; Paredes, A.H.; Frias, J.P.; Younes, Z.; Trotter, J.F.; Gunn, N.T.; Moussa, S.E.; et al. Efficacy and Safety of Aldafermin, an Engineered FGF19 Analog, in a Randomized, Double-Blind, Placebo-Controlled Trial of Patients with Nonalcoholic Steatohepatitis. Gastroenterology 2021, 160, 219–231.e1. [Google Scholar] [CrossRef]
  104. Gilroy, C.A.; Capozzi, M.E.; Varanko, A.K.; Tong, J.; D’Alessio, D.A.; Campbell, J.E.; Chilkoti, A. Sustained release of a GLP-1 and FGF21 dual agonist from an injectable depot protects mice from obesity and hyperglycemia. Sci. Adv. 2020, 6, eaaz9890. [Google Scholar] [CrossRef] [PubMed]
  105. Pan, Q.; Lin, S.; Li, Y.; Liu, L.; Li, X.; Gao, X.; Yan, J.; Gu, B.; Chen, X.; Li, W.; et al. A novel GLP-1 and FGF21 dual agonist has therapeutic potential for diabetes and non-alcoholic steatohepatitis. eBioMedicine 2021, 63, 103202. [Google Scholar] [CrossRef] [PubMed]
  106. Tschöp, M.H.; Finan, B.; Clemmensen, C.; Gelfanov, V.; Perez-Tilve, D.; Müller, T.D.; DiMarchi, R.D. Unimolecular Polypharmacy for Treatment of Diabetes and Obesity. Cell Metab. 2016, 24, 51–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Phase 3 Global Registration Trial. Available online: https://www.prnewswire.com/news-releases/lillys-tirzepatide-delivered-up-to-22-5-weight-loss-in-adults-with-obesity-or-overweight-in-surmount-1--301534871.html (accessed on 29 September 2022).
  108. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. Declaración PRISMA 2020: Una guía actualizada para la publicación de revisiones sistemáticas. Rev. Española Cardiol. 2021, 74, 790–799. [Google Scholar] [CrossRef]
  109. Staiger, H.; Keuper, M.; Berti, L.; de Angelis, M.H.; Häring, H.-U. Fibroblast Growth Factor 21—Metabolic Role in Mice and Men. Endocr. Rev. 2017, 38, 468–488. [Google Scholar] [CrossRef]
Figure 1. Description of the peripheral actions elicited by FGF21. FGF21 is a hepatic signal that improves several metabolic parameters such as energy expenditure, BAT thermogenesis, lipolysis, and glucose homeostasis. Abbreviations used: BAT: brown adipose tissue; FGF21: fibroblast growth factor-21; PPARg: Peroxisome proliferator-activated receptor gamma; WAT: white adipose tissue.
Figure 1. Description of the peripheral actions elicited by FGF21. FGF21 is a hepatic signal that improves several metabolic parameters such as energy expenditure, BAT thermogenesis, lipolysis, and glucose homeostasis. Abbreviations used: BAT: brown adipose tissue; FGF21: fibroblast growth factor-21; PPARg: Peroxisome proliferator-activated receptor gamma; WAT: white adipose tissue.
Ijms 23 13318 g001
Figure 2. FGF21/β-Klotho interactions in the CNS in metabolic control: FGF21 required β-klotho-containing receptor complexes in neurons to decrease body weight and circulating glucose and insulin concentrations, to induce energy expenditure and to signal WAT and BAT by SNS activation. Abbreviations used: CNS: central nervous system; BAT: brown adipose tissue; FGF21: fibroblast growth factor-21; Vglut2: vesicular glutamate transporter 2; WAT: white adipose tissue.
Figure 2. FGF21/β-Klotho interactions in the CNS in metabolic control: FGF21 required β-klotho-containing receptor complexes in neurons to decrease body weight and circulating glucose and insulin concentrations, to induce energy expenditure and to signal WAT and BAT by SNS activation. Abbreviations used: CNS: central nervous system; BAT: brown adipose tissue; FGF21: fibroblast growth factor-21; Vglut2: vesicular glutamate transporter 2; WAT: white adipose tissue.
Ijms 23 13318 g002
Figure 3. Description of the metabolic actions elicited by FGF21 in the hypothalamus. FGF21. FGF21 is a hepatic signal that induces food intake and improves several metabolic parameters such as energy expenditure, BAT thermogenesis, mainly by its action in the hypothalamus. CNS. Abbreviations used: Agrp: agouti related peptide; ARC: arcuate nucleus; GABA: gamma aminobutyric acid; DMH: dorsomedial hypothalamus; FGF21: fibroblast growth factor-21; LHA: lateral hypothalamic area; PVN: paraventricular hypothalamus; NPY: neuropeptide Y; Vglut2: vesicular glutamate transporter 2; VMH: ventromedial hypothalamus; SNS: sympathetic nervous system.
Figure 3. Description of the metabolic actions elicited by FGF21 in the hypothalamus. FGF21. FGF21 is a hepatic signal that induces food intake and improves several metabolic parameters such as energy expenditure, BAT thermogenesis, mainly by its action in the hypothalamus. CNS. Abbreviations used: Agrp: agouti related peptide; ARC: arcuate nucleus; GABA: gamma aminobutyric acid; DMH: dorsomedial hypothalamus; FGF21: fibroblast growth factor-21; LHA: lateral hypothalamic area; PVN: paraventricular hypothalamus; NPY: neuropeptide Y; Vglut2: vesicular glutamate transporter 2; VMH: ventromedial hypothalamus; SNS: sympathetic nervous system.
Ijms 23 13318 g003
Figure 4. Implications of FGF21 in the mesolimbic system: Abbreviations used: DA: dopamine; NAc: nucleus accumbens. FGF21: fibroblast growth factor-21; VTA: ventral tegmental area.
Figure 4. Implications of FGF21 in the mesolimbic system: Abbreviations used: DA: dopamine; NAc: nucleus accumbens. FGF21: fibroblast growth factor-21; VTA: ventral tegmental area.
Ijms 23 13318 g004
Figure 5. Flowchart of the study. ([“fibroblast growth factor 21” and “obesity”], [“fibroblast growth factor 21” and “feeding behavior”], [“fibroblast growth factor 21” and “liver”], [“fibroblast growth factor 21” and “brain”], [“fibroblast growth factor 21” and “Diabetes Mellitus, Type 2”], [“fibroblast growth factor 21” and “Non-alcoholic Fatty Liver Disease”], [“fibroblast growth factor 21” and “adipose Tissue, Brown”], [“fibroblast growth factor 21” and “Thermogenesis”], [“fibroblast growth factor 21” and “blood glucose”], [“fibroblast growth factor 21” and “insulin resistance”], [“fibroblast growth factor 21” and “humans”]).
Figure 5. Flowchart of the study. ([“fibroblast growth factor 21” and “obesity”], [“fibroblast growth factor 21” and “feeding behavior”], [“fibroblast growth factor 21” and “liver”], [“fibroblast growth factor 21” and “brain”], [“fibroblast growth factor 21” and “Diabetes Mellitus, Type 2”], [“fibroblast growth factor 21” and “Non-alcoholic Fatty Liver Disease”], [“fibroblast growth factor 21” and “adipose Tissue, Brown”], [“fibroblast growth factor 21” and “Thermogenesis”], [“fibroblast growth factor 21” and “blood glucose”], [“fibroblast growth factor 21” and “insulin resistance”], [“fibroblast growth factor 21” and “humans”]).
Ijms 23 13318 g005
Table 1. Metabolic actions of FGF21 mimetics or FGF21 signaling activation compounds: BW: body weight; db/db mice: mice lacking leptin receptor; DIO: diet induced obesity; EE: energy expenditure; FGF21: Fibroblast growth hormone 21; GLP-1: Glucagon like peptide-1; ICV: intracerebroventricular; ip: intraperitoneal; iv: intravenous; NAFLD: non-alcoholic fatty liver disease; NASH: non-alcoholic steatohepatitis; ob/ob mice: mice deficient in leptin; sc: subcutaneous; TG: triglyceride.
Table 1. Metabolic actions of FGF21 mimetics or FGF21 signaling activation compounds: BW: body weight; db/db mice: mice lacking leptin receptor; DIO: diet induced obesity; EE: energy expenditure; FGF21: Fibroblast growth hormone 21; GLP-1: Glucagon like peptide-1; ICV: intracerebroventricular; ip: intraperitoneal; iv: intravenous; NAFLD: non-alcoholic fatty liver disease; NASH: non-alcoholic steatohepatitis; ob/ob mice: mice deficient in leptin; sc: subcutaneous; TG: triglyceride.
StudySpecieAdministrationCompoundEffect
2020; Gilroy [104]Wild type micePeripheralGLP-1 and FGF21 dual agonistPotently reduces BW Decrease fasting glucose
Improves NASH
2021; Pan [105]Ob/ob mice
Db/db mice
Peripheral SCGLP-1 and FGF21 dual agonistPotently reduces BW
Improves glucose
Improve NAFLD
2008; Coskun [54]Ob/ob and DIO micePeripheral IVrecombinant human FGF21Potently reduces BW
Increases EE
Improves glucose
Improves NAFLD
2005; Kharitonenkov [55]Ob/ob and db/db micePeripheral SChuman FGF21Reduces plasma glucose and TG
2009; Xu [57]Ob/ob and DIO micePeripheral IPrecombinant human FGF21Improves glucose tolerance and insulin sensitivity
2007; Kharitonenkov [58]Diabetic non human primatesPeripheral IV SCrecombinant human FGF21Mild reduction of BW
Reduces plasma glucose and insulin and improves lipid profile
2013; Adams [59]DIO MicePeripheral SCFGF21 analogue, LY2405319Reduction of BW
Reduces plasma glucose and insulin and improves lipid profile
2013; Holland [66]Ob/ob and DIO micePeripheral SCrecombinant murine FGF21Reduces plasma glucose and improves insulin sensitivity
Increases adiponectin and EE
2015; Douris [71]WT miceICVrecombinant murine FGF21Energy expenditure, thermogenesis, and “browning”
2014; Owen [72]WT MiceICVrecombinant murine FGF21Increases thermogenesis
2017; Lan [73]DIO micePeripheral IPFGF21 mimetic antibodyReduces BW, plasma glucose and insulin, and improves lipid profile
2022: Pena-Leon, [90]RatsICVrecombinant human FGF21Reduces BW
Increases thermogenesis
2013; Gaich [94]Obese and diabetic HumansPeripheral SCFGF21 analog LY2405319Reduces BW and plasma insulin and improves lipid profile
2017; Kim [96]Obese humansPeripheral IVPF-05231023Not changes in BW
Potently reduces TG
2019; Charles [97]Obese and diabetic humansPeripheral SCPegbelfermin (BMS-986036), PEGylated FGF21Non changes in BW
Decrease fasting glucose and insulin sensitivity Improves lipid profile
Improve fibrosis markers
2019; Sanyal [98]overweight or obese with NASHPeripheral SCPegbelfermin (BMS-986036), PEGylated FGF21Non changes in BW
Improves NASH
2020; Baruch [99]Non human primatesPeripheral IVbispecific anti-FGFR1/KLB agonist antibody BFKB8488AReduces BW
Reduction in preference for sweet taste and carbohydrate intake
2020; Baruch [99]overweight or obesePeripheral SCbispecific anti-FGFR1/KLB agonist antibody BFKB8488ATransient reduction in BW
Improve lipid profile
Decrease fasting insulin
2018;Harrison [100]NASH patientsPeripheral S.C.FGF19 analogue, NGM282Reduces BW
Improves NASH
2020;Harrison [101]NASH patientsPeripheral S.C.FGF19 analogue, NGM282Improves NASH
2021;Harrison [103]NASH patientsPeripheral S.C.FGF19 analogue NGM282No changes in BW
Reduced liver fat
2015; Talukdar [84]DIO mice
Monkeys
Peripheral S.C.
I.V.
recombinant human FGF21
PF-05231023
Supresses sweet preferences
2016; Von Holstein-Rathlou [82]MicePeripheral IP and SCrecombinant human FGF21Suppresses sugar intake and sweet taste preferences
Table 2. Quality synthesis: ANCOVA: analysis of covariance; ALT: alanine aminotransferase; AST: aspartate aminotransferase; BW: body weight; FGF21:Fibroblast growth hormone 21; HDL: high density lipoprotein; LDL: low density lipoprotein; LS: Least squares; NASH: non-alcoholic steatohepatitis; SE: standard errors; SD: mean difference mean; TG: triglycerides.
Table 2. Quality synthesis: ANCOVA: analysis of covariance; ALT: alanine aminotransferase; AST: aspartate aminotransferase; BW: body weight; FGF21:Fibroblast growth hormone 21; HDL: high density lipoprotein; LDL: low density lipoprotein; LS: Least squares; NASH: non-alcoholic steatohepatitis; SE: standard errors; SD: mean difference mean; TG: triglycerides.
StudyModelDataCompound
2013; Gaich [94]Mixed effect linear modelBW: LS mean change from baseline = −1.75 (0.65) p < 0.05LY2405319
2017; Kim [96]Mixed-effects model for repeated
measurements
TG: Placebo-adjusted least squares mean
90% CI −62.1, −24.6
PF-05231023
2019; Charles [97]Longitudinal repeated-
measures analysis model
Glucose: LS mean
Estimates SE, and two-sided 90% confidence intervals (90% CI −3.46 to 0.22)
Fibrosis markers: % of change compared with baseline
ALT—20%;
AST—8%
BMS-986036
2020; Baruch [99]Descriptive statistics.Mean and SD % change for baseline:
BW: 2.30 (0.25)
Mean % change for baseline:
Cardiometabolic parameters:
TG: −66%
HDL: +34%
LDL: −37%
Adiponectin: +250%
BFKB8488A
2019; Sanyal [98]Longitudinal repeated measures analysis and unstructured covariance matrix were used to represent
the correlation of the repeated measures within each
patient
Hepatic fat:
Adjusted mean absolute change compared with placebo
Mean and SE, and two-sided 90% confidence intervals
(−6.8% vs. −1.3%; p = 0.0004)
BMS-986036
2018; Harrison [100]ANCOVABW: LS SD, SE
(95% CI), −2.0 (0.9; 3.7
to −0.3) p = 0.023
Hepatic fat: LS means
With SE, 95% (−11.1
1.4, −13.9 to −8.3; p < 0.0001)
NGM282 (FGF19 analogue)
2020; Harrison [101]Sensitivity analysis Wilcoxon matched pairs signed rank testFibrosis scores: change
compared with baseline
(0.5; 0.9 to −0) p = 0.035
NGM282 (FGF19 analogue)
2021; Harrison
[103]
ANCOVALeast squares (LS)
Means with standard
errors (SE), 95%
(difference, −5.0%; 95% CI, −8.0% to −1.9%;
p = 0.002)
FGF21 analogue aldafermin
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Prida, E.; Álvarez-Delgado, S.; Pérez-Lois, R.; Soto-Tielas, M.; Estany-Gestal, A.; Fernø, J.; Seoane, L.M.; Quiñones, M.; Al-Massadi, O. Liver Brain Interactions: Focus on FGF21 a Systematic Review. Int. J. Mol. Sci. 2022, 23, 13318. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232113318

AMA Style

Prida E, Álvarez-Delgado S, Pérez-Lois R, Soto-Tielas M, Estany-Gestal A, Fernø J, Seoane LM, Quiñones M, Al-Massadi O. Liver Brain Interactions: Focus on FGF21 a Systematic Review. International Journal of Molecular Sciences. 2022; 23(21):13318. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232113318

Chicago/Turabian Style

Prida, Eva, Sara Álvarez-Delgado, Raquel Pérez-Lois, Mateo Soto-Tielas, Ana Estany-Gestal, Johan Fernø, Luisa María Seoane, Mar Quiñones, and Omar Al-Massadi. 2022. "Liver Brain Interactions: Focus on FGF21 a Systematic Review" International Journal of Molecular Sciences 23, no. 21: 13318. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232113318

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop