Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Vaccines (Basel) ; 11(9)2023 Aug 25.
Article in English | MEDLINE | ID: mdl-37766096

ABSTRACT

Influenza vaccines are designed to mimic natural influenza virus exposure and stimulate a long-lasting immune response to future infections. The evolving nature of the influenza virus makes vaccination an important and efficacious strategy to reduce healthcare-related complications of influenza. Several lines of evidence indicate that influenza vaccination may induce nonspecific effects, also referred to as heterologous or pleiotropic effects, that go beyond protection against infection. Different explanations are proposed, including the upregulation and downregulation of cytokines and epigenetic reprogramming in monocytes and natural killer cells, imprinting an immunological memory in the innate immune system, a phenomenon termed "trained immunity". Also, cross-reactivity between related stimuli and bystander activation, which entails activation of B and T lymphocytes without specific recognition of antigens, may play a role. In this review, we will discuss the possible nonspecific effects of influenza vaccination in cardiovascular disease, type 1 diabetes, cancer, and Alzheimer's disease, future research questions, and potential implications. A discussion of the potential effects on infections by other pathogens is beyond the scope of this review.

2.
J Clin Endocrinol Metab ; 108(3): 653-664, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36240323

ABSTRACT

CONTEXT: Exogenous ketone body administration lowers circulating glucose levels but the underlying mechanisms are uncertain. OBJECTIVE: We tested the hypothesis that administration of the ketone body ß-hydroxybutyrate (ßOHB) acutely increases insulin sensitivity via feedback suppression of circulating free fatty acid (FFA) levels. METHODS: In a randomized, single-blinded crossover design, 8 healthy men were studied twice with a growth hormone (GH) infusion to induce lipolysis in combination with infusion of either ßOHB or saline. Each study day comprised a basal period and a hyperinsulinemic-euglycemic clamp combined with a glucose tracer and adipose tissue and skeletal muscle biopsies. RESULTS: ßOHB administration profoundly suppressed FFA levels concomitantly with a significant increase in glucose disposal and energy expenditure. This was accompanied by a many-fold increase in skeletal muscle content of both ßOHB and its derivative acetoacetate. CONCLUSION: Our data unravel an insulin-sensitizing effect of ßOHB, which we suggest is mediated by concomitant suppression of lipolysis.


Subject(s)
Human Growth Hormone , Insulin Resistance , Ketone Bodies , Humans , Male , 3-Hydroxybutyric Acid/pharmacology , Fatty Acids, Nonesterified , Glucose , Glucose Clamp Technique , Growth Hormone , Human Growth Hormone/pharmacology , Insulin/pharmacology , Insulin Resistance/physiology , Ketone Bodies/pharmacology , Ketone Bodies/therapeutic use , Lipolysis/drug effects , Lipolysis/physiology
3.
EBioMedicine ; 75: 103763, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34929488

ABSTRACT

BACKGROUND: Patients with active acromegaly exhibit insulin resistance despite a lean phenotype whereas controlled disease improves insulin sensitivity and increases fat mass. The mechanisms underlying this paradox remain elusive, but growth hormone (GH)-induced lipolysis plays a central role. The aim of the study was to investigative the molecular mechanisms of insulin resistance dissociated from obesity in patients with acromegaly. METHODS: In a prospective study, twenty-one patients with newly diagnosed acromegaly were studied at diagnosis and after disease control obtained by either surgery alone (n=10) or somatostatin analogue (SA) treatment (n=11) with assessment of body composition (DXA scan), whole body and tissue-specific insulin sensitivity and GH and insulin signalling in adipose tissue and skeletal muscle. FINDINGS: Disease control of acromegaly significantly reduced lean body mass (p<0.001) and increased fat mass (p<0.001). At diagnosis, GH signalling (pSTAT5) was constitutively activated in fat and enhanced expression of GH-regulated genes (CISH and IGF-I) were detected in muscle and fat. Insulin sensitivity in skeletal muscle, liver and adipose tissue increased after disease control regardless of treatment modality. This was associated with enhanced insulin signalling in both muscle and fat including downregulation of phosphatase and tensin homolog (PTEN) together with reduced signalling of GH and lipolytic activators in fat. INTERPRETATION: In conclusion, the study support that uncontrolled lipolysis is a major feature of insulin resistance in active acromegaly, and is characterized by upregulation of PTEN and suppression of insulin signalling in both muscle and fat. FUNDING: This work was supported by a grant from the Independent Research Fund, Denmark (7016-00303A) and from the Alfred Benzon Foundation, Denmark.


Subject(s)
Acromegaly , Insulin Resistance , Metabolic Syndrome , Acromegaly/complications , Acromegaly/metabolism , Adipose Tissue/metabolism , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Metabolic Syndrome/complications , Metabolic Syndrome/metabolism , Muscle, Skeletal/metabolism , Prospective Studies
4.
Ugeskr Laeger ; 183(21)2021 05 24.
Article in Danish | MEDLINE | ID: mdl-34060465

ABSTRACT

This case report describes a 57-year-old male with symptoms of tardive akathisia after long-term metoclopramide treatment. As metoclopramide is a dopamine receptor antagonist, it has the potential to cause drug-induced movement disorders, including akathisia, which is characterised by an inner restlessness resulting in a need for constant movement. Tardive akathisia, in contrast to acute akathisia, evolves after prolonged exposure to the triggering medication and can be a permanent condition. Treatment duration of metoclopramide should be restricted, and awareness of neurological side effects is important.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Metoclopramide , Akathisia, Drug-Induced/etiology , Dopamine Antagonists/adverse effects , Humans , Male , Metoclopramide/adverse effects , Middle Aged , Psychomotor Agitation
5.
Metabolism ; 105: 154188, 2020 04.
Article in English | MEDLINE | ID: mdl-32084431

ABSTRACT

OBJECTIVES: Lipoprotein lipase (LPL) catalyzes the hydrolysis of circulating triglycerides into free fatty acids (FFA) and thereby promotes FFA uptake in peripheral tissues. LPL is negatively regulated by angiopoietin-like protein 4 (ANGPTL4) presumably by an FFA-dependent mechanism. Growth hormone (GH) suppresses LPL activity, but it is unknown whether this is mediated by FFA and ANGPTL4. Therefore, we investigated the concerted effect of GH on ANGPTL4 and LPL in the presence and absence of lipolysis in two in vivo studies in human subjects. METHODS: In a randomized, placebo-controlled, cross-over study, nine obese men were examined after injection of 1) a GH bolus, and 2) a GH-receptor antagonist followed by four adipose tissue biopsies obtained over a 5-h period. In a second study, nine hypopituitary men were examined in a 2 × 2 factorial design including GH and acipimox (an anti-lipolytic agent), with biopsies from adipose tissue and skeletal muscle obtained during a basal period and a subsequent hyperinsulinemic-euglycemic clamp. The mRNA expression of ANGPTL4 and LPL as well as LPL activity were analyzed in the biopsies. RESULTS: In both studies, GH increased serum FFA levels, upregulated ANGPTL4 mRNA expression and suppressed LPL activity. In study 2, acipimox completely suppressed FFA levels and antagonized the effects of GH on ANGPTL4 and LPL. CONCLUSIONS: These human in vivo studies demonstrate that GH upregulates ANGPTL4 mRNA and suppresses LPL activity via an FFA-dependent mechanism.


Subject(s)
Angiopoietin-Like Protein 4/biosynthesis , Fatty Acids/metabolism , Human Growth Hormone/pharmacology , Lipoprotein Lipase/antagonists & inhibitors , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adult , Aged , Cross-Over Studies , Fatty Acids, Nonesterified/blood , Human Growth Hormone/antagonists & inhibitors , Humans , Hypolipidemic Agents/therapeutic use , Hypopituitarism/drug therapy , Hypopituitarism/metabolism , Male , Middle Aged , Obesity/metabolism , Obesity/pathology , Pyrazines/therapeutic use , RNA, Messenger/biosynthesis , Single-Blind Method , Up-Regulation/drug effects , Young Adult
6.
Mol Metab ; 29: 65-75, 2019 11.
Article in English | MEDLINE | ID: mdl-31668393

ABSTRACT

OBJECTIVE: Growth hormone (GH) stimulates lipolysis, but the underlying mechanisms remain incompletely understood. We examined the effect of GH on the expression of lipolytic regulators in adipose tissue (AT). METHODS: In a randomized, placebo-controlled, cross-over study, nine men were examined after injection of 1) a GH bolus and 2) a GH-receptor antagonist (pegvisomant) followed by four AT biopsies. In a second study, eight men were examined in a 2 × 2 factorial design including GH infusion and 36-h fasting with AT biopsies obtained during a basal period and a hyperinsulinemic-euglycemic clamp. Expression of GH-signaling intermediates and lipolytic regulators were studied by PCR and western blotting. In addition, mechanistic experiments in mouse models and 3T3-L1 adipocytes were performed. RESULTS: The GH bolus increased circulating free fatty acids (p < 0.0001) together with phosphorylation of signal transducer and activator of transcription 5 (STAT5) (p < 0.0001) and mRNA expression of the STAT5-dependent genes cytokine-inducible SH2-containing protein (CISH) and IGF-1 in AT. This was accompanied by suppressed mRNA expression of G0/G1 switch gene 2 (G0S2) (p = 0.007) and fat specific protein 27 (FSP27) (p = 0.002) and upregulation of phosphatase and tensin homolog (PTEN) mRNA expression (p = 0.03). Suppression of G0S2 was also observed in humans after GH infusion and fasting, as well as in GH transgene mice, and in vitro studies suggested MEK-PPARγ signaling to be involved. CONCLUSIONS: GH-induced lipolysis in human subjects in vivo is linked to downregulation of G0S2 and FSP27 and upregulation of PTEN in AT. Mechanistically, in vitro data suggest that GH acts via MEK to suppress PPARγ-dependent transcription of G0S2. ClinicalTrials.govNCT02782221 and NCT01209429.


Subject(s)
Adipose Tissue/metabolism , Human Growth Hormone/analogs & derivatives , Human Growth Hormone/administration & dosage , Adipose Tissue/pathology , Adult , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Biomarkers/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cross-Over Studies , Down-Regulation/drug effects , Fatty Acids, Nonesterified/blood , Human Growth Hormone/pharmacology , Humans , Lipolysis , Male , Mice , Mice, Transgenic , PPAR gamma/metabolism , Placebo Effect , Signal Transduction , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...