Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Mol Metab ; 51: 101259, 2021 09.
Article in English | MEDLINE | ID: mdl-34033941

ABSTRACT

OBJECTIVE: Skeletal muscle is an attractive target for blood glucose-lowering pharmacological interventions. Oral dosing of small molecule direct pan-activators of AMPK that bind to the allosteric drug and metabolite (ADaM) site, lowers blood glucose through effects in skeletal muscle. The molecular mechanisms responsible for this effect are not described in detail. This study aimed to illuminate the mechanisms by which ADaM-site activators of AMPK increase glucose uptake in skeletal muscle. Further, we investigated the consequence of co-stimulating muscles with two types of AMPK activators i.e., ADaM-site binding small molecules and the prodrug AICAR. METHODS: The effect of the ADaM-site binding small molecules (PF739 and 991), AICAR or co-stimulation with PF739 or 991 and AICAR on muscle glucose uptake was investigated ex vivo in m. extensor digitorum longus (EDL) excised from muscle-specific AMPKα1α2 as well as whole-body AMPKγ3-deficient mouse models. In vitro complex-specific AMPK activity was measured by immunoprecipitation and molecular signaling was assessed by western blotting in muscle lysate. To investigate the transferability of these studies, we treated diet-induced obese mice in vivo with PF739 and measured complex-specific AMPK activation in skeletal muscle. RESULTS: Incubation of skeletal muscle with PF739 or 991 increased skeletal muscle glucose uptake in a dose-dependent manner. Co-incubating PF739 or 991 with a maximal dose of AICAR increased glucose uptake to a greater extent than any of the treatments alone. Neither PF739 nor 991 increased AMPKα2ß2γ3 activity to the same extent as AICAR, while co-incubation led to potentiated effects on AMPKα2ß2γ3 activation. In muscle from AMPKγ3 KO mice, AICAR-stimulated glucose uptake was ablated. In contrast, the effect of PF739 or 991 on glucose uptake was not different between WT and AMPKγ3 KO muscles. In vivo PF739 treatment lowered blood glucose levels and increased muscle AMPKγ1-complex activity 2-fold, while AMPKα2ß2γ3 activity was not affected. CONCLUSIONS: ADaM-site binding AMPK activators increase glucose uptake independently of AMPKγ3. Co-incubation with PF739 or 991 and AICAR potentiates the effects on muscle glucose uptake and AMPK activation. In vivo, PF739 lowers blood glucose and selectively activates muscle AMPKγ1-complexes. Collectively, this suggests that pharmacological activation of AMPKγ1-containing complexes in skeletal muscle can increase glucose uptake and can lead to blood glucose lowering.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Blood Glucose/drug effects , Muscle, Skeletal/metabolism , Obesity/drug therapy , Ribonucleotides/pharmacology , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/pharmacology , Aminoimidazole Carboxamide/therapeutic use , Animals , Blood Glucose/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Humans , Mice , Mice, Knockout , Muscle, Skeletal/drug effects , Obesity/blood , Obesity/etiology , Obesity/metabolism , Phosphorylation/drug effects , Ribonucleotides/therapeutic use , Signal Transduction/drug effects
2.
Cell Death Dis ; 12(6): 506, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34006821

ABSTRACT

Diabetes (DB) is a risk factor for osteoarthritis progression. High glucose (HG) is one of the key pathological features of DB and has been demonstrated to induce apoptosis and senescence in chondrocytes. Autophagy is an endogenous mechanism that can protect cells against apoptosis and senescence. The effects of HG on autophagy in cells including chondrocytes have been studied; however, the results have been inconsistent. The current study aimed to elucidate the underlying mechanisms, which could be associated with the contrasting outcomes. The present study revealed that HG can induce apoptosis and senescence in chondrocytes, in addition to regulating autophagy dynamically. The present study demonstrated that HG can cause oxidative stress in chondrocytes and suppress the AMPK pathway in a dose-dependent manner. Elimination of oxidative stress by Acetylcysteine, also called N-acetyl cysteine (NAC), downregulated autophagy and alleviated HG-stimulated apoptosis and senescence, while activation of the AMPK signaling pathway by AICAR not only upregulated autophagy but also alleviated HG-stimulated apoptosis and senescence. A combined treatment of NAC and AICAR was superior to treatment with either NAC or AICAR. The study has demonstrated that HG can suppress autophagy through the AMPK pathway and induce autophagy via oxidative stress in chondrocytes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Acetylcysteine/therapeutic use , Aminoimidazole Carboxamide/analogs & derivatives , Autophagy/physiology , Chondrocytes/metabolism , Diabetes Complications/complications , Diabetes Mellitus/drug therapy , Glucose/metabolism , Hyperglycemia/complications , Oxidative Stress/physiology , Ribonucleotides/therapeutic use , Acetylcysteine/pharmacology , Aminoimidazole Carboxamide/pharmacology , Aminoimidazole Carboxamide/therapeutic use , Animals , Humans , Mice , Ribonucleotides/pharmacology
3.
Biomed Pharmacother ; 139: 111579, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33845375

ABSTRACT

Alzheimer's disease (AD) is the most common type of dementia worldwide, characterized by the deposition of neurofibrillary tangles and amyloid-ß (Aß) peptides in the brain. Additionally, increasing evidence demonstrates that a neuroinflammatory state and oxidative stress, iron-dependent, play a crucial role in the onset and disease progression. Besides conventional therapies, the use of natural-based products represents a future medical option for AD treatment and/or prevention. We, therefore, evaluated the effects of a ribonucleotides-based ingredient (Ribodiet®) in a non-genetic mouse model of AD. To this aim, mice were injected intracerebroventricularly (i.c.v.) with Aß1-42 peptide (3 µg/3 µl) and after with Ribodiet® (0.1-10 mg/mouse) orally (p.o.) 3 times weekly for 21 days following the induction of experimental AD. The mnemonic and cognitive decline was then evaluated, and, successively, we have assessed ex vivo the modulation of different cyto-chemokines on mice brain homogenates. Finally, the level of GFAP, S100ß, and iron-related metabolic proteins were monitored as markers of reactive gliosis, neuro-inflammation, and oxidative stress. Results indicate that Ribodiet® lessens oxidative stress, brain inflammation, and amyloid pathology via modulation of iron-related metabolic proteins paving the way for its rationale use for the treatment of AD and other age-related diseases.


Subject(s)
Alzheimer Disease/prevention & control , Cerebral Amyloid Angiopathy/prevention & control , Dietary Supplements , Encephalitis/prevention & control , Oxidative Stress/drug effects , Ribonucleotides/therapeutic use , Alzheimer Disease/psychology , Amyloid beta-Peptides , Animals , Behavior, Animal/drug effects , Biomarkers , Cerebral Amyloid Angiopathy/psychology , Diet , Encephalitis/psychology , Gliosis/prevention & control , Injections, Intraventricular , Male , Mice , Nonheme Iron Proteins/metabolism , Peptide Fragments , Psychomotor Performance/drug effects , Ribonucleotides/pharmacology
4.
Zhonghua Jie He He Hu Xi Za Zhi ; 43(3): 173-176, 2020 Mar 12.
Article in Chinese | MEDLINE | ID: mdl-32164081

ABSTRACT

The new coronavirus pneumonia (NCP), also named as COVID-19 by WHO on Feb 11 2020, is now causing a severe public health emergency in China since. The number of diagnosed cases is more than 40,000 until the submission of this manuscript. Coronavirus has caused several epidemic situations world widely, but the present contagious disease caused by 2019 new coronavirus is unprecedentedly fulminating. The published cohorts of 2019 new coronavirus (n-Cov) are single-center studies, or retrospective studies. We here share the therapeutic experiences of NCP treatment with literature review. Combination of Ribavirin and interferon-α is recommended by the 5(th) edition National Health Commission's Regimen (Revised Edition) because of the effect on Middle East respiratory syndrome (MERS), and the effectiveness of Lopinavir/Ritonavir and Remdisivir needs to be confirmed by randomized controlled trial (RCT), given the situation of no specific antivirus drug on NCP is unavailable. Systemic glucocorticosteroid is recommended as a short term use (1~2 mg·kg(-1)·d(-1), 3~5 d) by the 5(th) edition National Health Commission's Regimen (Revised Edition) yet RCTs are expected to confirm the effectiveness. Inappropriate application of antibiotics should be avoided, especially the combination of broad-spectrum antibiotics, for the NCP is not often complicated with bacterial infection.


Subject(s)
Adenosine/analogs & derivatives , Alanine/analogs & derivatives , Antiviral Agents , Coronavirus Infections/drug therapy , Cytochrome P-450 CYP3A Inhibitors/therapeutic use , Lopinavir/therapeutic use , Pneumonia, Viral/drug therapy , Ribonucleotides/therapeutic use , Ritonavir/therapeutic use , Adenosine/therapeutic use , Adenosine Monophosphate/analogs & derivatives , Alanine/therapeutic use , Anti-Bacterial Agents/therapeutic use , COVID-19 , China , Drug Therapy, Combination , Humans , Retrospective Studies
5.
FASEB J ; 34(4): 5016-5026, 2020 04.
Article in English | MEDLINE | ID: mdl-32090372

ABSTRACT

Progression of blinding diseases, such as age-related macular degeneration, is accelerated by light exposure. However, no particular intervention is applied to the photostress. Here, we report neuroprotective effects of the adenosine monophosphate (AMP)-activated protein kinase (AMPK) activator, 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR), on light-induced visual function impairment, photoreceptor disorders and death in mice. Increase in retinal ATP levels in response to photostress was transient, because oxygen consumption rate (OCR) and cytochrome c oxidase (CcO) activity were reduced under photostress. However, AICAR treatment preserved OCR, CcO activity, and high levels of retinal ATP after light exposure. AMPK knockdown in the photoreceptor-derived cell line revealed that AMPK targeted CcO activity. Further, our data indicated that photostress reduced mitochondrial respiratory function and ATP levels, while AICAR treatment promoted neuronal survival and retained visual function, stabilizing ATP levels through preserved CcO activity. The current study has provided proof of concept for providing cells with sufficient energy to promote cell survival in the presence of cellular stress. This is in contrast to the previous reports which primarily investigated therapeutic approaches to suppress stress signals. Hence, stabilization of the ATP supply may serve as a novel therapeutic approach to support tissue survival under stress and prevent neurodegeneration.


Subject(s)
Adenosine Triphosphate/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Macular Degeneration/drug therapy , Neuroprotective Agents/pharmacology , Protein Kinases/metabolism , Ribonucleotides/pharmacology , AMP-Activated Protein Kinase Kinases , Aminoimidazole Carboxamide/pharmacology , Aminoimidazole Carboxamide/therapeutic use , Animals , Cell Line , Electron Transport Complex IV/metabolism , Macular Degeneration/etiology , Macular Degeneration/metabolism , Male , Mice , Mice, Inbred BALB C , Neuroprotective Agents/therapeutic use , Oxygen Consumption , Protein Kinases/genetics , Retina/drug effects , Retina/metabolism , Retina/radiation effects , Ribonucleotides/therapeutic use , Ultraviolet Rays/adverse effects
6.
Cardiovasc Drugs Ther ; 34(2): 255-269, 2020 04.
Article in English | MEDLINE | ID: mdl-32034646

ABSTRACT

Doxorubicin is a commonly used chemotherapeutic agent for the treatment of a range of cancers, but despite its success in improving cancer survival rates, doxorubicin is cardiotoxic and can lead to congestive heart failure. Therapeutic options for this patient group are limited to standard heart failure medications with the only drug specific for doxorubicin cardiotoxicity to reach FDA approval being dexrazoxane, an iron-chelating agent targeting oxidative stress. However, dexrazoxane has failed to live up to its expectations from preclinical studies while also bringing up concerns about its safety. Despite decades of research, the molecular mechanisms of doxorubicin cardiotoxicity are still poorly understood and oxidative stress is no longer considered to be the sole evil. Mitochondrial impairment, increased apoptosis, dysregulated autophagy and increased fibrosis have also been shown to be crucial players in doxorubicin cardiotoxicity. These cellular processes are all linked by one highly conserved intracellular kinase: adenosine monophosphate-activated protein kinase (AMPK). AMPK regulates mitochondrial biogenesis via PGC1α signalling, increases oxidative mitochondrial metabolism, decreases apoptosis through inhibition of mTOR signalling, increases autophagy through ULK1 and decreases fibrosis through inhibition of TGFß signalling. AMPK therefore sits at the control point of many mechanisms shown to be involved in doxorubicin cardiotoxicity and cardiac AMPK signalling itself has been shown to be impaired by doxorubicin. In this review, we introduce different agents known to activate AMPK (metformin, statins, resveratrol, thiazolidinediones, AICAR, specific AMPK activators) as well as exercise and dietary restriction, and we discuss the existing evidence for their potential role in cardioprotection from doxorubicin cardiotoxicity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antibiotics, Antineoplastic/adverse effects , Doxorubicin/adverse effects , Enzyme Activators/therapeutic use , Heart Diseases/prevention & control , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/therapeutic use , Animals , Caloric Restriction , Cardiotoxicity , Enzyme Activation , Exercise , Heart Diseases/chemically induced , Heart Diseases/enzymology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Metformin/pharmacology , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Resveratrol/therapeutic use , Ribonucleotides/therapeutic use , Signal Transduction , Thiazolidinediones/therapeutic use
7.
Stem Cell Res Ther ; 11(1): 45, 2020 02 03.
Article in English | MEDLINE | ID: mdl-32014016

ABSTRACT

BACKGROUND: Mesenchymal stromal cell (MSC) stemness capacity diminishes over prolonged in vitro culture, which negatively affects their application in regenerative medicine. To slow down the senescence of MSCs, here, we have evaluated the in vitro effects of 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), an AMPK activator, and nicotinamide (NAM), an activator of sirtuin1 (SIRT1). METHODS: Human adipose-derived MSCs were cultured to passage (P) 5. Subsequently, the cells were grown in either normal medium alone (control group), the medium supplemented with AICAR (1 mM) and NAM (5 mM), or in the presence of both for 5 weeks to P10. Cell proliferation, differentiation capacity, level of apoptosis and autophagy, morphological changes, total cellular reactive oxygen species (ROS), and activity of mTORC1 and AMPK were compared among different treatment groups. RESULTS: MSCs treated with AICAR, NAM, or both displayed an increase in proliferation and osteogenic differentiation, which was augmented in the group receiving both. Treatment with AICAR or NAM led to decreased expression of ß-galactosidase, reduced accumulation of dysfunctional lysosomes, and characteristic morphologic features of young MSCs. Furthermore, while NAM administration could significantly reduce the total cellular ROS in aged MSCs, AICAR treatment did not. Moreover, AICAR-treated cells possess a high proliferation capacity; however, they also show the highest level of cellular apoptosis. The observed effects of AICAR and NAM were in light of the attenuated mTORC1 activity and increased AMPK activity and autophagy. CONCLUSIONS: Selective inhibition of mTORC1 by AICAR and NAM boosts autophagy, retains MSCs' self-renewal and multi-lineage differentiation capacity, and postpones senescence-associated changes after prolonged in vitro culture. Additionally, co-administration of AICAR and NAM shows an additive or probably a synergistic effect on cellular senescence.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Hypoglycemic Agents/therapeutic use , Mesenchymal Stem Cells/drug effects , Niacinamide/therapeutic use , Ribonucleotides/therapeutic use , Vitamin B Complex/therapeutic use , Aminoimidazole Carboxamide/pharmacology , Aminoimidazole Carboxamide/therapeutic use , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Humans , Hypoglycemic Agents/pharmacology , Niacinamide/pharmacology , Ribonucleotides/pharmacology , Vitamin B Complex/pharmacology
8.
Biosci Trends ; 14(1): 69-71, 2020 Mar 16.
Article in English | MEDLINE | ID: mdl-31996494

ABSTRACT

As of January 22, 2020, a total of 571 cases of the 2019-new coronavirus (2019-nCoV) have been reported in 25 provinces (districts and cities) in China. At present, there is no vaccine or antiviral treatment for human and animal coronavirus, so that identifying the drug treatment options as soon as possible is critical for the response to the 2019-nCoV outbreak. Three general methods, which include existing broad-spectrum antiviral drugs using standard assays, screening of a chemical library containing many existing compounds or databases, and the redevelopment of new specific drugs based on the genome and biophysical understanding of individual coronaviruses, are used to discover the potential antiviral treatment of human pathogen coronavirus. Lopinavir /Ritonavir, Nucleoside analogues, Neuraminidase inhibitors, Remdesivir, peptide (EK1), abidol, RNA synthesis inhibitors (such as TDF, 3TC), anti-inflammatory drugs (such as hormones and other molecules), Chinese traditional medicine, such ShuFengJieDu Capsules and Lianhuaqingwen Capsule, could be the drug treatment options for 2019-nCoV. However, the efficacy and safety of these drugs for 2019- nCoV still need to be further confirmed by clinical experiments.


Subject(s)
Antiviral Agents/therapeutic use , Betacoronavirus , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Alanine/therapeutic use , COVID-19 , Drug Combinations , Drug Discovery , Humans , Lopinavir/therapeutic use , Neuraminidase/antagonists & inhibitors , Nucleosides/therapeutic use , Ribonucleotides/therapeutic use , Ritonavir/therapeutic use , SARS-CoV-2 , COVID-19 Drug Treatment
9.
Curr Protein Pept Sci ; 21(1): 66-77, 2020.
Article in English | MEDLINE | ID: mdl-31424367

ABSTRACT

Alzheimer's disease (AD) is the most common neurodegenerative disorder. The pathogenesis of AD is very complicated. For decades, the amyloid hypothesis has influenced and guided research in the field of AD. Meanwhile, researchers gradually realized that AD is caused by multiple concomitant factors, such as autophagy, mitochondrial quality control, insulin resistance and oxidative stress. In current clinical trials, the improvement strategies of AD, such as Aß antibody immunotherapy and gamma secretase inhibitors, are limited. There is mounting evidence of neurodegenerative disorders indicated that activation of AMP-activated protein kinase (AMPK) may have broad neuroprotective effects. We reviewed the researches on AMPK for AD, the results demonstrated that activation of AMPK is controversial in Aß deposition and tau phosphorylation, but is positive to promote autophagy, maintain mitochondrial quality control, reduce insulin resistance and relieve oxidative stress. It is concluded that AMPK might be a new target for AD by aggressively treating the risk factors in the future.


Subject(s)
AMP-Activated Protein Kinases/genetics , Alzheimer Disease/drug therapy , Aminoimidazole Carboxamide/analogs & derivatives , Amyloid beta-Peptides/metabolism , Neuroprotective Agents/therapeutic use , Pyrones/therapeutic use , Ribonucleotides/therapeutic use , Thiophenes/therapeutic use , tau Proteins/genetics , AMP-Activated Protein Kinases/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Aminoimidazole Carboxamide/therapeutic use , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Autophagy/drug effects , Autophagy/genetics , Biphenyl Compounds , Clinical Trials as Topic , Gene Expression Regulation , Humans , Insulin Resistance , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress/drug effects , Phosphorylation , tau Proteins/metabolism
10.
N Engl J Med ; 381(24): 2293-2303, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31774950

ABSTRACT

BACKGROUND: Although several experimental therapeutics for Ebola virus disease (EVD) have been developed, the safety and efficacy of the most promising therapies need to be assessed in the context of a randomized, controlled trial. METHODS: We conducted a trial of four investigational therapies for EVD in the Democratic Republic of Congo, where an outbreak began in August 2018. Patients of any age who had a positive result for Ebola virus RNA on reverse-transcriptase-polymerase-chain-reaction assay were enrolled. All patients received standard care and were randomly assigned in a 1:1:1:1 ratio to intravenous administration of the triple monoclonal antibody ZMapp (the control group), the antiviral agent remdesivir, the single monoclonal antibody MAb114, or the triple monoclonal antibody REGN-EB3. The REGN-EB3 group was added in a later version of the protocol, so data from these patients were compared with those of patients in the ZMapp group who were enrolled at or after the time the REGN-EB3 group was added (the ZMapp subgroup). The primary end point was death at 28 days. RESULTS: A total of 681 patients were enrolled from November 20, 2018, to August 9, 2019, at which time the data and safety monitoring board recommended that patients be assigned only to the MAb114 and REGN-EB3 groups for the remainder of the trial; the recommendation was based on the results of an interim analysis that showed superiority of these groups to ZMapp and remdesivir with respect to mortality. At 28 days, death had occurred in 61 of 174 patients (35.1%) in the MAb114 group, as compared with 84 of 169 (49.7%) in the ZMapp group (P = 0.007), and in 52 of 155 (33.5%) in the REGN-EB3 group, as compared with 79 of 154 (51.3%) in the ZMapp subgroup (P = 0.002). A shorter duration of symptoms before admission and lower baseline values for viral load and for serum creatinine and aminotransferase levels each correlated with improved survival. Four serious adverse events were judged to be potentially related to the trial drugs. CONCLUSIONS: Both MAb114 and REGN-EB3 were superior to ZMapp in reducing mortality from EVD. Scientifically and ethically sound clinical research can be conducted during disease outbreaks and can help inform the outbreak response. (Funded by the National Institute of Allergy and Infectious Diseases and others; PALM ClinicalTrials.gov number, NCT03719586.).


Subject(s)
Alanine/analogs & derivatives , Antibodies, Monoclonal/therapeutic use , Antiviral Agents/therapeutic use , Hemorrhagic Fever, Ebola/drug therapy , Ribonucleotides/therapeutic use , Adenosine Monophosphate/analogs & derivatives , Adolescent , Adult , Alanine/adverse effects , Alanine/therapeutic use , Antibodies, Monoclonal/adverse effects , Antiviral Agents/adverse effects , Child , Child, Preschool , Democratic Republic of the Congo/epidemiology , Disease Outbreaks , Ebolavirus/genetics , Female , Hemorrhagic Fever, Ebola/mortality , Humans , Infant , Infant, Newborn , Infusions, Intravenous , Male , RNA, Viral/blood , Ribonucleotides/adverse effects , Single-Blind Method , Young Adult
11.
Int J Mol Sci ; 20(13)2019 Jul 02.
Article in English | MEDLINE | ID: mdl-31269729

ABSTRACT

The stimulation of the AMP-activated kinase (AMPK) by 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide (AICAR) has been associated with antihyperalgesia and the inhibition of nociceptive signaling in the spinal cord in models of paw inflammation. The attenuated nociception comes along with a strongly reduced paw edema, indicating that peripheral antiinflammatory mechanisms contribute to antinociception. In this study, we investigated the impact of AICAR on the immune cell composition in inflamed paws, as well as the regulation of inflammatory and resolving markers in macrophages. By using fluorescence-activated cell sorting (FACS) analysis and immunofluorescence, we found a significantly increased fraction of proresolving M2 macrophages and anti-inflammatory interleukin (IL)-10 in inflamed tissue, while M1 macrophages and proinflammatory cytokines such as IL-1 were decreased by AICAR in wild type mice. In AMPKα2 knock-out mice, the M2 polarization of macrophages in the paw was missing. The results were supported by experiments in primary macrophage cultures which also showed a shift to a proresolving phenotype with decreased levels of proinflammatory mediators and increased levels of antiinflammatory mediators. However, in the cell cultures, we did not observe differences between the AMPKα2+/+ and -/- cells, thus indicating that the AICAR-induced effects are at least partially AMPK-independent. In summary, our results indicate that AICAR has potent antiinflammatory and proresolving properties in inflammation which are contributing to a reduction of inflammatory edema and antinociception.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Macrophages/drug effects , Ribonucleotides/therapeutic use , Aminoimidazole Carboxamide/therapeutic use , Animals , Cells, Cultured , Edema/complications , Edema/drug therapy , Edema/immunology , Hyperalgesia/complications , Hyperalgesia/drug therapy , Hyperalgesia/immunology , Inflammation/complications , Inflammation/immunology , Macrophages/immunology , Male , Mice, Inbred C57BL
12.
J Neuroinflammation ; 16(1): 34, 2019 Feb 12.
Article in English | MEDLINE | ID: mdl-30755236

ABSTRACT

BACKGROUND: Chronic pain is a major clinical problem with limited treatment options. Previous studies have demonstrated that activation of adenosine monophosphate-activated protein kinase (AMPK) can attenuate neuropathic pain. Inflammation/immune response at the site of complete Freund's adjuvant (CFA) injection is known to be a critical trigger of the pathological changes that produce inflammatory pain. However, whether activation of AMPK produces an analgesic effect through inhibiting the proinflammatory cytokines, including interleukin-1ß (IL-1ß), in inflammatory pain remains unknown. METHODS: Inflammatory pain was induced in mice injected with CFA. The effects of AICAR (5-aminoimidazole-4-carboxyamide ribonucleoside, an AMPK activator), Compound C (an AMPK inhibitor), and IL-1ra (an IL-1 receptor antagonist) were tested at day 4 after CFA injection. Inflammatory pain was assessed with von Frey filaments and hot plate. Immunoblotting, hematoxylin and eosin (H&E) staining, and immunofluorescence were used to assess inflammation-induced biochemical changes. RESULTS: The AMPK activator AICAR produced an analgesic effect and inhibited the level of proinflammatory cytokine IL-1ß in the inflamed skin in mice. Moreover, activation of AMPK suppressed CFA-induced NF-κB p65 translocation from the cytosol to the nucleus in activated macrophages (CD68+ and CX3CR1+) of inflamed skin tissues. Subcutaneous injection of IL-1ra attenuated CFA-induced inflammatory pain. The AMPK inhibitor Compound C and AMPKα shRNA reversed the analgesic effect of AICAR and the effects of AICAR on IL-1ß and NF-κB activation in inflamed skin tissues. CONCLUSIONS: Our study provides new information that AMPK activation produces the analgesic effect by inhibiting NF-κB activation and reducing the expression of IL-1ß in inflammatory pain.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Hypoglycemic Agents/therapeutic use , Interleukin-1beta/metabolism , NF-kappa B/metabolism , Pain/metabolism , Ribonucleotides/therapeutic use , Aminoimidazole Carboxamide/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Activation/physiology , Freund's Adjuvant/toxicity , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Inflammation/chemically induced , Inflammation/complications , Inflammation/drug therapy , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pain/drug therapy , Pain/etiology , Pain Threshold/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Skin/drug effects , Skin/metabolism , Skin/pathology
13.
J Mol Med (Berl) ; 97(3): 423-434, 2019 03.
Article in English | MEDLINE | ID: mdl-30721324

ABSTRACT

Recent studies have indicated that the Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) pathway is closely involved in liver fibrosis and other fibrotic diseases. However, whether targeting the AMPK pathway can rescue liver fibrosis and its complications, such as portal hypertension, is unknown. This study aimed to explore the therapeutic value of AICAR (5-aminoimidazole-4-carboxyamide ribonucleoside), an agonist of the AMPK pathway, on liver fibrosis and portal hypertension in bile duct ligation (BDL) rats. In vitro experiments showed that the gene expression levels of TGF-b, a-SMA, and collagen 1 in primary rat hepatic stellate cells (HSCs) were significantly decreased after AICAR treatment. The p-eNOS expression and nitric oxide (NO) production were increased by AICAR administration in sinusoidal endothelial cells (SECs). For in vivo animal studies, AICAR acutely decreased portal pressure in the BDL and CCL4 fibrotic rats, but not in the partial portal vein ligation (PVL) rats, without changes in systemic hemodynamics. It was also observed by using intravital fluorescence microscopy that AICAR led to sinusoidal vasodilation in situ experiment. We propose that the relevant mechanisms may be related to the activation of the AMPK/NO pathway in SECs and that this activation promoted NO production in the liver, thereby promoting hepatic sinusoid microcirculation and decreased intrahepatic resistance. The results were verified using the NO inhibitor L-NAME. Chronic AICAR treatment also showed profound beneficial effects on the BDL model rats. The hemodynamic condition was greatly improved, but the positive effect could be partially blocked by L-NAME. Moreover, AICAR also decreased hepatic fibrogenesis in the BDL rats. KEY MESSAGES: Acute and chronic use of AICAR could alleviate portal pressure without changing systemic hemodynamics. AICAR induced sinusoidal vasodilation by improving NO bioavailability and ameliorating endothelial dysfunction in vivo and in vitro. AICAR could alleviate liver cirrhosis in the BDL model rats.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Hypertension, Portal/drug therapy , Liver Cirrhosis/drug therapy , Nitric Oxide/metabolism , Ribonucleotides/pharmacology , Ribonucleotides/therapeutic use , Actins/genetics , Actins/metabolism , Aminoimidazole Carboxamide/pharmacology , Aminoimidazole Carboxamide/therapeutic use , Animals , Bile Ducts/surgery , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hypertension, Portal/genetics , Hypertension, Portal/metabolism , Hypertension, Portal/physiopathology , Ligation , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/physiopathology , Male , Nitric Oxide Synthase Type III/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transforming Growth Factor beta/genetics
15.
Biochem Biophys Res Commun ; 509(3): 680-686, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30616891

ABSTRACT

Cisplatin causes acute kidney injury (AKI) through proximal tubular injury. We investigated the protective effect of the adenosine monophosphate protein kinase (AMPK) activator 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) against cisplatin-induced AKI. We investigated whether the AMP-kinase activator AICAR ameliorates cisplatin-induced AKI through the JAK/STAT/SOCS pathway. Male Sprague-Dawley (SD) rats were randomly divided into four groups: control, AICAR, cisplatin, and cisplatin + AICAR. As appropriate to their treatment group, the rats were injected with a single dose of cisplatin (7 mg/kg, i.p.). AICAR was administered to the rats at 100 mg/kg i.p. daily. Blood urea nitrogen (BUN) and serum creatinine were measured. Renal damage was analyzed in sections stained with hematoxylin and eosin (H&E). Renal tissues were also examined by immunohistochemistry and western blot for p-AMPK, Kim-1, cleaved caspase 3, and JAK/STAT/SOCS. For in vitro studies, NRK-52E normal rat kidney cells were treated with cisplatin and/or AICAR. By western blot, we confirmed the expression of p-AMPK and the JAK/STAT/SOCS pathway in NRK-52E cells. AICAR was protective against cisplatin-induced acute tubular injury by up-regulating p-AMPK expression in NRK-52E cells. Protein expression levels of JAK2/STAT1 were markedly ameliorated in NRK-52E cells by AICAR. The protective mechanism of AICAR may be associated with suppression of the JAK2/STAT1 pathway and up-regulation of SOCS1, an inhibitor of the JAK2/STAT1 pathway. The present study demonstrates the protective effects of AICAR against cisplatin-induced AKI and shows a new renoprotective mechanism through the JAK2/STAT1/SOCS1 pathway and apoptosis inhibition. This study suggests that activation of the AMPK activator AICAR might ameliorate cisplatin-induced AKI.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/prevention & control , Aminoimidazole Carboxamide/analogs & derivatives , Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Enzyme Activators/therapeutic use , Ribonucleotides/therapeutic use , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Aminoimidazole Carboxamide/therapeutic use , Animals , Cell Line , Janus Kinases/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Rats, Sprague-Dawley , STAT Transcription Factors/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism
17.
Eur J Pharmacol ; 833: 100-108, 2018 Aug 15.
Article in English | MEDLINE | ID: mdl-29856969

ABSTRACT

This study aims to investigate the effect of κ-opioid receptor activation on myocardial ischemia and reperfusion(I/R) injury and elucidate the underlying mechanisms. Myocardial I/R rat model and simulated I/R cardiomyocytes model were established. In vivo study showed that U50,488 H improved cardiac function, reduced myocardial infarct size and serum cTnT significantly. The effect of U50,488 H was abolished by nor-BNI(a κ-opioid receptor antagonist), Compound C(an AMPK inhibitor), Akt inhibitor and L-NAME(an eNOS inhibitor). AICAR, an AMPK activator, mimicked the effect of U50,488 H. U50,488 H up-regulated p-AMPK, p-Akt, and p-eNOS, which were abolished by nor-BNI. AICAR increased p-Akt and p-eNOS, which was abolished by Compound C. In vitro study showed that U50,488 H increased p-AMPK, p-Akt, and p-eNOS via κ-OR activation. The effect of U50,488 H on p-AMPK was abolished by compound C, but not Akt inhibitor and L-NAME. The effect of U50,488 H on p-Akt was abolished by compound C and Akt inhibitor, but not L-NAME. AICAR increased p-Akt and p-eNOS, which was abolished by Akt inhibitor, but not L-NAME. U50,488 H and AICAR also increased the viability of cardiomyocytes subjected to simulated I/R, the effects of U50,488 H and AICAR were blocked by nor-BNI, Compound C, Akt inhibitor, and L-NAME, respectively. In conclusion, κ-OR activation confers cardioprotection via AMPK/Akt/eNOS signaling.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Myocardial Reperfusion Injury/metabolism , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Opioid, kappa/metabolism , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/therapeutic use , AMP-Activated Protein Kinases/antagonists & inhibitors , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Aminoimidazole Carboxamide/therapeutic use , Animals , Cell Line , Cell Survival/drug effects , Male , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Rats, Sprague-Dawley , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/antagonists & inhibitors , Ribonucleotides/pharmacology , Ribonucleotides/therapeutic use , Signal Transduction , Troponin T/blood
19.
EMBO Mol Med ; 10(7)2018 07.
Article in English | MEDLINE | ID: mdl-29844217

ABSTRACT

Activation of AMPK has been associated with pro-atrophic signaling in muscle. However, AMPK also has anti-inflammatory effects, suggesting that in cachexia, a syndrome of inflammatory-driven muscle wasting, AMPK activation could be beneficial. Here we show that the AMPK agonist AICAR suppresses IFNγ/TNFα-induced atrophy, while the mitochondrial inhibitor metformin does not. IFNγ/TNFα impair mitochondrial oxidative respiration in myotubes and promote a metabolic shift to aerobic glycolysis, similarly to metformin. In contrast, AICAR partially restored metabolic function. The effects of AICAR were prevented by the AMPK inhibitor Compound C and were reproduced with A-769662, a specific AMPK activator. AICAR and A-769662 co-treatment was found to be synergistic, suggesting that the anti-cachectic effects of these drugs are mediated through AMPK activation. AICAR spared muscle mass in mouse models of cancer and LPS induced atrophy. Together, our findings suggest a dual function for AMPK during inflammation-driven atrophy, wherein it can play a protective role when activated exogenously early in disease progression, but may contribute to anabolic suppression and atrophy when activated later through mitochondrial dysfunction and subsequent metabolic stress.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Cachexia/prevention & control , Metformin/therapeutic use , Protein Kinases/metabolism , Ribonucleotides/therapeutic use , AMP-Activated Protein Kinase Kinases , Aminoimidazole Carboxamide/therapeutic use , Animals , Cachexia/etiology , Cell Line , Enzyme Activation , Inflammation/complications , Interferon-gamma/antagonists & inhibitors , Male , Mice, Inbred BALB C , Mitochondria/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Neoplasms, Experimental/pathology , Nitric Oxide Synthase Type II/metabolism , Protein Kinases/drug effects , Shock, Septic/chemically induced , Shock, Septic/complications , Tumor Necrosis Factor-alpha/antagonists & inhibitors
20.
Invest New Drugs ; 36(5): 763-772, 2018 10.
Article in English | MEDLINE | ID: mdl-29504069

ABSTRACT

Breast cancer has been emerging as a most common threat among women, thus many efforts were made to find drugs for fighting breast cancer. So far, PI3K (Phosphatidylinositol-4,5-bisphosphate 3-kinase) inhibitors have been believed to be effective drugs until frequent resistance emerged. Recently, PI3K H1047R mutation has been reported to sensitize breast cancer cells to PI3K inhibition by aspirin. Considering aspirin activates AMPK (AMP-activated protein kinase) simultaneously, it is possible that AMPK activators and PI3K inhibitors can synergistically inhibit breast cancers. Here we clearly observed synergistic suppression of cell growth in all three breast cancer cell lines (MCF-7, MDA-MB-361 and HCC38) when co-treating cells with PI3K inhibitor GDC-0941 and AMPK activator AICAR (5-Aminoimidazole-4-carboxamide ribonucleotide). What is more, it is rather remarkable that the synergistic effect was much more dramatic in PIK3CA (PI3K catalytic subunit alpha) mutated (E545K) cells (MCF-7 and MDA-MB-361) than in PIK3CA wild-type cells (HCC38), which implied there is a relationship between PI3K genetic status and the efficacy of combination therapy. By using PIK3CA wild-type isogenic MCF-7 cell line, which exhibited attenuated cell proliferation compared with the parental MCF-7 cell line, we found endogenous reverse mutation of PIK3CA E545K alleles to wild-type sequence in MCF-7 cells dramatically impaired the synergy of PI3Ki&Ka (combinatorial PI3K inhibition and AMPK activation). Furthermore, PI3Ki&Ka significantly attenuated tumorigenesis of parental MCF-7 cells but not PIK3CA wild-type isogenic MCF-7 cells in tumor xenograft models. Taken together, our results suggest a promising precision therapy of PI3Ki&Ka in PIK3CA mutant breast cancers.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Class I Phosphatidylinositol 3-Kinases/genetics , Indazoles/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Ribonucleotides/therapeutic use , Sulfonamides/therapeutic use , Aminoimidazole Carboxamide/pharmacology , Aminoimidazole Carboxamide/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Drug Synergism , Humans , Indazoles/pharmacology , Mice, Inbred BALB C , Mice, Nude , Mutation , Protein Kinase Inhibitors/pharmacology , Ribonucleotides/pharmacology , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...