Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (45)

Search Parameters:
Keywords = AMPK-STAT3 signaling pathway

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
29 pages, 2859 KB  
Article
α-Linolenic Acid Alleviates Diabetic Cardiomyopathy by Activating AMPK-STAT3 Pathway to Inhibit Ferritinophagy and Enhance SLC7A11-GPX4 Antioxidant Axis
by Ziqian Zhang, Xue Bai, Qian Du and Jianhong Yang
Molecules 2026, 31(1), 79; https://doi.org/10.3390/molecules31010079 - 24 Dec 2025
Abstract
Diabetic cardiomyopathy (DCM) is a severe complication of diabetes, in which ferroptosis is a key pathogenic mechanism. This study examines how alpha-linolenic acid (ALA), a plant-derived omega-3 polyunsaturated fatty acid, protects against damage from ferroptosis in DCM. Using an in vitro model of [...] Read more.
Diabetic cardiomyopathy (DCM) is a severe complication of diabetes, in which ferroptosis is a key pathogenic mechanism. This study examines how alpha-linolenic acid (ALA), a plant-derived omega-3 polyunsaturated fatty acid, protects against damage from ferroptosis in DCM. Using an in vitro model of H9C2 cardiomyocytes treated with high glucose/palmitate, combined with a high-fat diet and mouse model of low-dose streptozotocin (STZ)-induced diabetes, this research demonstrates for the first time that ALA significantly alleviates cardiac dysfunction and prevents ferroptosis. Mechanistically, ALA inhibits STAT3 phosphorylation by activating the AMPK signaling pathway, thereby reducing NCOA4-mediated ferritinophagy and mitigating mitochondrial iron overload and reactive oxygen species accumulation. It also enhances the function of the SLC7A11/GSH/GPX4 axis, reducing lipid peroxidation (LPO)-induced ferroptosis. Collectively, these findings indicate that ALA protects against diabetic cardiomyopathy by coordinating the regulation of ferritinophagy and antioxidant defense through the AMPK-STAT3 pathway, offering a potential therapeutic strategy for disease management. Full article
25 pages, 1376 KB  
Review
Mollugin: A Comprehensive Review of Its Multifaceted Pharmacological Properties and Therapeutic Potential
by Sandra Ross Olakkengil Shajan, Bushra Zia, Charu Sharma, Sandeep B. Subramanya and Shreesh Ojha
Int. J. Mol. Sci. 2025, 26(24), 12003; https://doi.org/10.3390/ijms262412003 - 13 Dec 2025
Viewed by 239
Abstract
The substantial interest in plant-based drugs or plant-derived phytocompounds drives researchers to conduct comprehensive investigations on their therapeutic properties. Mollugin, one of the major active constituents of Rubia cardifolia, has been well-studied for its pharmacological properties, demonstrating potent anti-inflammatory properties by suppressing [...] Read more.
The substantial interest in plant-based drugs or plant-derived phytocompounds drives researchers to conduct comprehensive investigations on their therapeutic properties. Mollugin, one of the major active constituents of Rubia cardifolia, has been well-studied for its pharmacological properties, demonstrating potent anti-inflammatory properties by suppressing the TAK-1-mediated activation of NF-κB/MAPK and enhancing the Nrf2/HO-1-mediated antioxidant response. It exhibits strong anticancer effects through ferroptosis via IGF2BP3/GPX4 pathways, induces mitochondrial apoptosis, and targets NF-κB, ERK, and PI3K/Akt/mTOR to suppress tumor progression. Mollugin also inhibits JAK2/STAT and PARP1 pathways, suppressing IL-1β expression via the modulation of ZFP91. Moreover, it regulates the MAPK/p38 pathway, promotes neuroprotection, and improves cognitive performance through GLP-1 receptor activation. Mollugin promotes osteogenesis by activating the BMP-2/Smad1/5/8 signaling pathway and downregulates MAPK, Akt, and GSK3β expression, leading to the inhibition of osteoclastogenesis. It overcomes multidrug resistance by downregulating MDR1/P-gp, CREB, NF-κB, and COX-2 through AMPK activation. Its antibacterial effect is mediated by strong binding to FUR, UDP, and IpxB proteins in Enterobacter xiangfangensis. Mollugin mitigates Klebsiella pneumoniae infection, suppresses adipogenesis without causing cytotoxicity, and protects endothelial cells via the BDNF/TrkB-Akt signaling pathway. Synthetic derivatives of mollugin, such as oxomollugin and azamollugin, have shown enhanced anticancer and anti-inflammatory effects by regulating EGFR, PKM2, TLR4/MyD88/IRAK/TRAF6, and NF-κB/IRF3 pathways with improved solubility and stability. Collectively, these findings emphasize the broad-spectrum activity of mollugin. This review provides a critical interpretation of the mechanistic pathways regulated by mollugin and its derivatives, emphasizing their pharmacological significance and exploring their potential for future translation as multitarget drug candidates. Full article
(This article belongs to the Special Issue Plant-Derived Bioactive Compounds for Pharmacological Applications)
Show Figures

Graphical abstract

29 pages, 3722 KB  
Review
Glial Cells in the Early Stages of Neurodegeneration: Pathogenesis and Therapeutic Targets
by Eugenia Ahremenko, Alexander Andreev, Danila Apushkin and Eduard Korkotian
Int. J. Mol. Sci. 2025, 26(24), 11995; https://doi.org/10.3390/ijms262411995 - 12 Dec 2025
Viewed by 441
Abstract
Astrocytes and microglia constitute nearly half of all central nervous system cells and are indispensable for its proper function. Both exhibit striking morphological and functional heterogeneity, adopting either neuroprotective (A2, M2) or proinflammatory (A1, M1) phenotypes in response to cytokines, pathogen-associated molecular patterns [...] Read more.
Astrocytes and microglia constitute nearly half of all central nervous system cells and are indispensable for its proper function. Both exhibit striking morphological and functional heterogeneity, adopting either neuroprotective (A2, M2) or proinflammatory (A1, M1) phenotypes in response to cytokines, pathogen-associated molecular patterns (PAMPs)/damage-associated molecular patterns (DAMPs), toll-like receptor 4 (TLR4) activation, and NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome signaling. Crucially, many of these phenotypic transitions arise during the earliest stages of neurodegeneration, when glial dysfunction precedes overt neuronal loss and may act as a primary driver of disease onset. This review critically examines glial-centered hypotheses of neurodegeneration, with emphasis on their roles in early disease phases: (i) microglial polarization from an M2 neuroprotective state to an M1 proinflammatory state; (ii) NLRP3 inflammasome assembly via P2X purinergic receptor 7 (P2X7R)-mediated K+ efflux; (iii) a self-amplifying astrocyte–microglia–neuron inflammatory feedback loop; (iv) impaired microglial phagocytosis and extracellular-vesicle–mediated propagation of β-amyloid (Aβ) and tau; (v) astrocytic scar formation driven by aquaporin-4 (AQP4), matrix metalloproteinase-9 (MMP-9), glial fibrillary acidic protein (GFAP)/vimentin, connexins, and janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) signaling; (vi) cellular reprogramming of astrocytes and NG2 glia into functional neurons; and (vii) mitochondrial dysfunction in glia, including Dynamin-related protein 1/Mitochondrial fission protein 1 (Drp1/Fis1) fission imbalance and dysregulation of the sirtuin 1/peroxisome proliferator-activated receptor gamma coactivator 1-alpha (Sirt1/PGC-1α) axis. Promising therapeutic strategies target pattern-recognition receptors (TLR4, NLRP3/caspase-1), cytokine modulators (interleukin-4 (IL-4), interleukin-10 (IL-10)), signaling cascades (JAK2–STAT, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), phosphoinositide 3-kinase–protein kinase B (PI3K–AKT), adenosine monophosphate-activated protein kinase (AMPK)), microglial receptors (triggering receptor expressed on myeloid cells 2 (TREM2)/spleen tyrosine kinase (SYK)/ DNAX-activating protein 10 (DAP10), siglec-3 (CD33), chemokine C-X3-C motif ligand 1/ CX3C motif chemokine receptor 1 (CX3CL1/CX3CR1), Cluster of Differentiation 200/ Cluster of Differentiation 200 receptor 1 (CD200/CD200R), P2X7R), and mitochondrial biogenesis pathways, with a focus on normalizing glial phenotypes rather than simply suppressing pathology. Interventions that restore neuroglial homeostasis at the earliest stages of disease may hold the greatest potential to delay or prevent progression. Given the complexity of glial phenotypes and molecular isoform diversity, a comprehensive, multitargeted approach is essential for mitigating Alzheimer’s disease and related neurodegenerative disorders. This review not only synthesizes pathogenesis but also highlights therapeutic opportunities, offering what we believe to be the first concise overview of the principal hypotheses implicating glial cells in neurodegeneration. Rather than focusing on isolated mechanisms, our goal is a holistic perspective—integrating diverse glial processes to enable comparison across interconnected pathological conditions. Full article
(This article belongs to the Special Issue Early Molecular Markers of Neurodegeneration)
Show Figures

Graphical abstract

18 pages, 641 KB  
Review
Coenzyme Q10 and Intracellular Signalling Pathways: Clinical Relevance
by David Mantle
Int. J. Mol. Sci. 2025, 26(22), 11024; https://doi.org/10.3390/ijms262211024 - 14 Nov 2025
Viewed by 1455
Abstract
Intracellular signalling pathways provide a mechanism to connect events at a cell surface to the nucleus and are of fundamental importance to normal cell functioning. Intracellular signalling pathways control many aspects of cell metabolism, including mitochondrial function, oxidative stress, inflammation, and apoptosis/ferroptosis. Randomised [...] Read more.
Intracellular signalling pathways provide a mechanism to connect events at a cell surface to the nucleus and are of fundamental importance to normal cell functioning. Intracellular signalling pathways control many aspects of cell metabolism, including mitochondrial function, oxidative stress, inflammation, and apoptosis/ferroptosis. Randomised controlled clinical trials supplementing coenzyme Q10 (CoQ10) have reported significant clinical improvements in a number of disorders, in turn associated with the action of CoQ10 to promote normal mitochondrial function, reduce oxidative stress and inflammation, and mediate apoptosis and ferroptosis. However, the precise mechanisms by which CoQ10 facilitates beneficial changes in the above factors is not completely understood. In the present article, the evidence we have reviewed provides a supporting rationale that the beneficial role of CoQ10 in the above disorders occurs via mediation of major intracellular signalling pathways, including the Nrf2/NQO1, NF-κB, P13/AKT/mTOR, MAPK, JAK/STAT, WNT/B-catenin, AMPK-YAP-OPA1, and hedgehog (Hh) pathways; the clinical consequences of such mediation are also reviewed. Full article
(This article belongs to the Special Issue Signalling Pathways in Metabolic Diseases and Cancers)
Show Figures

Figure 1

38 pages, 1248 KB  
Review
Targeting Inflammation with Natural Products: A Mechanistic Review of Iridoids from Bulgarian Medicinal Plants
by Rositsa Mihaylova, Viktoria Elincheva, Reneta Gevrenova, Dimitrina Zheleva-Dimitrova, Georgi Momekov and Rumyana Simeonova
Molecules 2025, 30(17), 3456; https://doi.org/10.3390/molecules30173456 - 22 Aug 2025
Cited by 2 | Viewed by 2276
Abstract
Chronic, low-grade inflammation is a key contributor to the development of numerous non-communicable diseases (NCDs), including cardiovascular, metabolic, and neurodegenerative disorders. Conventional anti-inflammatory drugs, such as nonsteroidal anti-inflammatory drugs (NSAIDs) and corticosteroids, often present safety concerns with prolonged use, highlighting the need for [...] Read more.
Chronic, low-grade inflammation is a key contributor to the development of numerous non-communicable diseases (NCDs), including cardiovascular, metabolic, and neurodegenerative disorders. Conventional anti-inflammatory drugs, such as nonsteroidal anti-inflammatory drugs (NSAIDs) and corticosteroids, often present safety concerns with prolonged use, highlighting the need for safer, multi-targeted therapeutic options. Iridoids, a class of monoterpenoid compounds abundant in several medicinal plants, have emerged as promising bioactive agents with diverse pharmacological properties. They exert anti-inflammatory and metabolic regulatory effects by modulating key signaling pathways, including nuclear factor kappa B (NF-κB), mitogen-activated protein kinase (MAPK), Janus kinase/signal transducer and activator of transcription (JAK/STAT), adenosine monophosphate-activated protein kinase (AMPK), and peroxisome proliferator-activated receptor (PPAR) pathways. This review provides a comprehensive summary of the major iridoid metabolites derived from ten Bulgarian medicinal plant species, along with mechanistic insights from in vitro and in vivo studies. Documented biological activities include anti-inflammatory, antioxidant, immunomodulatory, antifibrotic, organoprotective, antibacterial, antiviral, analgesic, and metabolic effects. By exploring their phytochemical profiles and pharmacodynamics, we underscore the therapeutic potential of iridoid-rich Bulgarian flora in managing inflammation-related and metabolic diseases. These findings support the relevance of iridoids as complementary or alternative agents to conventional therapies and highlight the need for further translational and clinical research. Full article
(This article belongs to the Special Issue Role of Natural Products in Inflammation)
Show Figures

Figure 1

28 pages, 2605 KB  
Review
Exercise-Induced Muscle–Fat Crosstalk: Molecular Mediators and Their Pharmacological Modulation for the Maintenance of Metabolic Flexibility in Aging
by Amelia Tero-Vescan, Hans Degens, Antonios Matsakas, Ruxandra Ștefănescu, Bianca Eugenia Ősz and Mark Slevin
Pharmaceuticals 2025, 18(8), 1222; https://doi.org/10.3390/ph18081222 - 19 Aug 2025
Cited by 1 | Viewed by 4788
Abstract
Regular physical activity induces a dynamic crosstalk between skeletal muscle and adipose tissue, modulating the key molecular pathways that underlie metabolic flexibility, mitochondrial function, and inflammation. This review highlights the role of myokines and adipokines—particularly IL-6, irisin, leptin, and adiponectin—in orchestrating muscle–adipose tissue [...] Read more.
Regular physical activity induces a dynamic crosstalk between skeletal muscle and adipose tissue, modulating the key molecular pathways that underlie metabolic flexibility, mitochondrial function, and inflammation. This review highlights the role of myokines and adipokines—particularly IL-6, irisin, leptin, and adiponectin—in orchestrating muscle–adipose tissue communication during exercise. Exercise stimulates AMPK, PGC-1α, and SIRT1 signaling, promoting mitochondrial biogenesis, fatty acid oxidation, and autophagy, while also regulating muscle hypertrophy through the PI3K/Akt/mTOR and Wnt/β-catenin pathways. Simultaneously, adipose-derived factors like leptin and adiponectin modulate skeletal muscle metabolism via JAK/STAT3 and AdipoR1-mediated AMPK activation. Additionally, emerging exercise mimetics such as the mitochondrial-derived peptide MOTS-c and myostatin inhibitors are highlighted for their roles in increasing muscle mass, the browning of white adipose tissue, and improving systemic metabolic function. The review also addresses the role of anti-inflammatory compounds, including omega-3 polyunsaturated fatty acids and low-dose aspirin, in mitigating NF-κB and IL-6 signaling to protect mitochondrial health. The resulting metabolic flexibility, defined as the ability to efficiently switch between lipid and glucose oxidation, is enhanced through repeated exercise, counteracting age- and disease-related mitochondrial and functional decline. Together, these adaptations demonstrate the importance of inter-tissue signaling in maintaining energy homeostasis and preventing sarcopenia, obesity, and insulin resistance. Finally, here we propose a stratified treatment algorithm based on common age-related comorbidities, offering a framework for precision-based interventions that may offer a promising strategy to preserve metabolic plasticity and delay the age-associated decline in cardiometabolic health. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

13 pages, 839 KB  
Review
Strategies of Classical Swine Fever Immune Evasion
by Yuanji Zhang, Fangtao Li and Yebing Liu
Int. J. Mol. Sci. 2025, 26(16), 7838; https://doi.org/10.3390/ijms26167838 - 14 Aug 2025
Viewed by 1267
Abstract
Classical swine fever (CSF) is a highly contagious and lethal disease caused by classical swine fever virus (CSFV), and it is also a notifiable disease according to the World Organization for Animal Health. Owing to the continuous growth of the international trade in [...] Read more.
Classical swine fever (CSF) is a highly contagious and lethal disease caused by classical swine fever virus (CSFV), and it is also a notifiable disease according to the World Organization for Animal Health. Owing to the continuous growth of the international trade in pigs and pig products, pig farming has become the pillar industry of the global livestock industry and is the most important source of animal protein for mankind. As a single-stranded RNA virus, CSFV can avoid being recognized and cleared by the host immune system through a variety of immune evasion strategies so that it persists in the host body and causes multisystemic pathology. CSF has also become one of the most serious infectious diseases affecting the pig industry, resulting in considerable economic losses to the pig industry. Therefore, understanding the main immune evasion mechanism of CSFV is very important for the prevention and control of CSF infection. This article reviews the main immune evasion mechanisms of CSFV, including the suppression of nonspecific immune responses; evasion of adaptive immune responses; and the regulation of host cell apoptosis and cell autophagy. CSFV affects type I interferon regulatory signals; the JAK-STAT signaling pathway; the RIG-I and NF-κB signaling pathways; immune cell function; the mitochondrial apoptosis pathway; and the endoplasmic reticulum stress apoptosis pathway; the PI3K-Akt signaling mediated AMPK-mTOR macroautophagy pathway through its structural proteins Erns and E1 and E2; and the nonstructural proteins Npro, NS4B, and NS5A to achieve immune evasion. As our understanding of CSFV immune strategies continues to deepen, we believe that this understanding will provide new strategies for the development of new vaccines and novel diagnostic methods in the future. Full article
(This article belongs to the Special Issue Immune Responses to Viruses)
Show Figures

Figure 1

31 pages, 2599 KB  
Review
Therapeutic Potential of Ginsenosides in Anthracycline-Induced Cardiotoxicity
by Rongrong Bai, Zhigao Zhao, Xing Han, Mingying Shang, Guangxue Liu, Feng Xu and Shaoqing Cai
Molecules 2025, 30(12), 2527; https://doi.org/10.3390/molecules30122527 - 10 Jun 2025
Viewed by 2189
Abstract
Anthracyclines play an irreplaceable role in cancer treatment, although their clinical application is limited due to severe side effects such as arrhythmia, cardiomyopathy, and myocardial infarction. The currently available clinical drugs for treating anthracycline-induced cardiotoxicity (AIC) are limited by numerous drawbacks, including the [...] Read more.
Anthracyclines play an irreplaceable role in cancer treatment, although their clinical application is limited due to severe side effects such as arrhythmia, cardiomyopathy, and myocardial infarction. The currently available clinical drugs for treating anthracycline-induced cardiotoxicity (AIC) are limited by numerous drawbacks, including the side effects of the therapeutic agents, single treatment mechanisms, and individual patient variations. Therefore, novel drugs with broader applicability and multitarget synergistic protective effects are, therefore, urgently needed. Ginsenosides, the primary bioactive constituents of plants belonging to the genus Panax (family Araliaceae), exhibit a wide range of pharmacological activities, including anti-inflammatory, antioxidative, and antitumor effects, and have demonstrated cardioprotective properties against AIC. This article examines the mechanisms of AIC and the modulatory effects of ginsenosides on these mechanisms. This review highlights the potential molecular targets and signaling pathways through which ginsenosides exert therapeutic effects on AIC, including the regulation of oxidative-stress-related pathways such as Keap1/Nrf2, MAPK, STAT, PI3K/Akt, and AMPK; the restoration of mitochondrial function; the modulation of autophagy; and the inhibition of pyroptosis, ferroptosis, and apoptosis. Therefore, this review serves as a theoretical basis and provides a research direction for future investigation regarding the prevention and treatment of AIC with ginsenosides, as well as clinical translation studies. Full article
Show Figures

Figure 1

24 pages, 2492 KB  
Review
Antioxidant Peptides Derived from Woody Oil Resources: Mechanisms of Redox Protection and Emerging Therapeutic Opportunities
by Jia Tu, Jie Peng, Li Wen, Changzhu Li, Zhihong Xiao, Ying Wu, Zhou Xu, Yuxi Hu, Yan Zhong, Yongjun Miao, Jingjing Xiao and Sisi Liu
Pharmaceuticals 2025, 18(6), 842; https://doi.org/10.3390/ph18060842 - 4 Jun 2025
Viewed by 1479
Abstract
Antioxidant peptides derived from woody oil resource by-products exhibit strong free radical scavenging abilities and offer potential applications in functional foods, nutraceuticals, and cosmetics. This review summarizes the latest advances in preparation technologies, including enzymatic hydrolysis, microbial fermentation, chemical synthesis, recombinant expression, and [...] Read more.
Antioxidant peptides derived from woody oil resource by-products exhibit strong free radical scavenging abilities and offer potential applications in functional foods, nutraceuticals, and cosmetics. This review summarizes the latest advances in preparation technologies, including enzymatic hydrolysis, microbial fermentation, chemical synthesis, recombinant expression, and molecular imprinting, each with distinct advantages in yield, selectivity, and scalability. The structure–activity relationships of antioxidant peptides are explored with respect to amino acid composition, molecular weight, and 3D conformation, which collectively determine their bioactivity and stability. Additionally, emerging delivery systems—such as nanoliposomes, microencapsulation, and cell-penetrating peptides—are discussed for their role in enhancing peptide stability, absorption, and targeted release. Mechanistic studies reveal that antioxidant peptides from woody oil resources act through network pharmacology, engaging core signaling pathways, including Nrf2/ARE, PI3K/Akt, AMPK, and JAK/STAT, to regulate oxidative stress, mitochondrial health, and inflammation. Preliminary safety data from in vitro, animal, and early clinical studies suggest low toxicity and favorable tolerability. The integration of omics technologies, molecular docking, and bioinformatics is accelerating the mechanism-driven design and functional validation of peptides. In conclusion, antioxidant peptides derived from woody oil resources represent a sustainable, multifunctional, and scalable solution for improving human health and promoting a circular bioeconomy. Future research should focus on structural optimization, delivery enhancement, and clinical validation to facilitate their industrial translation. Full article
Show Figures

Figure 1

55 pages, 2579 KB  
Review
Regulation and Function of Tumor-Associated Macrophages (TAMs) in Colorectal Cancer (CRC): The Role of the SRIF System in Macrophage Regulation
by Agnieszka Geltz, Jakub Geltz and Aldona Kasprzak
Int. J. Mol. Sci. 2025, 26(11), 5336; https://doi.org/10.3390/ijms26115336 - 1 Jun 2025
Cited by 3 | Viewed by 3735
Abstract
Colorectal cancer (CRC) remains the leading cause of morbidity and mortality for both men and women worldwide. Tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) of solid tumors, including CRC. These macrophages are found in the pro-inflammatory [...] Read more.
Colorectal cancer (CRC) remains the leading cause of morbidity and mortality for both men and women worldwide. Tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) of solid tumors, including CRC. These macrophages are found in the pro-inflammatory M1 and anti-inflammatory M2 forms, with the latter increasingly recognized for its tumor-promoting phenotypes. Many signaling molecules and pathways, including AMPK, EGFR, STAT3/6, mTOR, NF-κB, MAPK/ERK, and HIFs, are involved in regulating TAM polarization. Consequently, researchers are investigating several potential predictive and prognostic markers, and novel TAM-based therapeutic targets, especially in combination therapies for CRC. Macrophages of the gastrointestinal tract, including the normal colon and rectum, produce growth hormone-releasing inhibitory peptide/somatostatin (SRIF/SST) and five SST receptors (SSTRs, SST1-5). While the immunosuppressive function of the SRIF system is primarily known for various tissues, its role within CRC-associated TAMs remains underexplored. This review focuses on the following three aspects of TAMs: first, the role of macrophages in the normal colon and rectum within the broader context of macrophage biology; second, the various bioactive factors and signaling pathways associated with TAM function, along with potential strategies targeting TAMs in CRC; and third, the interaction between the SRIF system and macrophages in both normal tissues and the CRC microenvironment. Full article
(This article belongs to the Special Issue The Role of Macrophages in Cancers)
Show Figures

Figure 1

26 pages, 1674 KB  
Review
Branched-Chain Amino Acids and Inflammation Management in Endurance Sports: Molecular Mechanisms and Practical Implications
by Miaomiao Xu, Danting Hu, Xiaoguang Liu, Zhaowei Li and Liming Lu
Nutrients 2025, 17(8), 1335; https://doi.org/10.3390/nu17081335 - 12 Apr 2025
Cited by 3 | Viewed by 13237
Abstract
Endurance athletes frequently experience muscle damage and inflammation due to prolonged, high-intensity exercise, which can impair recovery and hinder performance. This review examines the role of branched-chain amino acid (BCAA) supplementation in muscle repair, inflammation modulation, and immune regulation. BCAAs—particularly leucine and isoleucine—activate [...] Read more.
Endurance athletes frequently experience muscle damage and inflammation due to prolonged, high-intensity exercise, which can impair recovery and hinder performance. This review examines the role of branched-chain amino acid (BCAA) supplementation in muscle repair, inflammation modulation, and immune regulation. BCAAs—particularly leucine and isoleucine—activate key molecular pathways, including the mechanistic target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK), to promote muscle protein synthesis and enhance energy metabolism. They also attenuate inflammatory responses by modulating the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), mitogen-activated protein kinase (MAPK), and Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways, reducing levels of tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). In addition, BCAAs influence immune function via mechanistic target of rapamycin complex 1 (mTORC1) signaling, enhance autophagy, and mitigate exercise-induced apoptosis. These molecular effects result in reduced muscle soreness, lower muscle damage biomarker levels (e.g., creatine kinase, lactate dehydrogenase), and improved recovery. Practical considerations such as optimal dosage, timing, and co-supplementation with carbohydrates, proteins, or omega-3s are also addressed. While BCAAs show promise as a nutritional strategy for enhancing recovery and controlling inflammation in endurance athletes, further research is needed to refine personalized protocols and clarify long-term effects. Full article
(This article belongs to the Section Proteins and Amino Acids)
Show Figures

Figure 1

30 pages, 2069 KB  
Review
Anti-Inflammatory Effects of SGLT2 Inhibitors: Focus on Macrophages
by Elena Y. Rykova, Vadim V. Klimontov, Elena Shmakova, Anton I. Korbut, Tatyana I. Merkulova and Julia Kzhyshkowska
Int. J. Mol. Sci. 2025, 26(4), 1670; https://doi.org/10.3390/ijms26041670 - 15 Feb 2025
Cited by 32 | Viewed by 7027
Abstract
A growing body of evidence indicates that nonglycemic effects of sodium–glucose cotransporter 2 (SGLT2) inhibitors play an important role in the protective effects of these drugs in diabetes, chronic kidney disease, and heart failure. In recent years, the anti-inflammatory potential of SGLT2 inhibitors [...] Read more.
A growing body of evidence indicates that nonglycemic effects of sodium–glucose cotransporter 2 (SGLT2) inhibitors play an important role in the protective effects of these drugs in diabetes, chronic kidney disease, and heart failure. In recent years, the anti-inflammatory potential of SGLT2 inhibitors has been actively studied. This review summarizes results of clinical and experimental studies on the anti-inflammatory activity of SGLT2 inhibitors, with a special focus on their effects on macrophages, key drivers of metabolic inflammation. In patients with type 2 diabetes, therapy with SGLT2 inhibitors reduces levels of inflammatory mediators. In diabetic and non-diabetic animal models, SGLT2 inhibitors control low-grade inflammation by suppressing inflammatory activation of tissue macrophages, recruitment of monocytes from the bloodstream, and macrophage polarization towards the M1 phenotype. The molecular mechanisms of the effects of SGLT2 inhibitors on macrophages include an attenuation of inflammasome activity and inhibition of the TLR4/NF-κB pathway, as well as modulation of other signaling pathways (AMPK, PI3K/Akt, ERK 1/2-MAPK, and JAKs/STAT). The review discusses the state-of-the-art concepts and prospects of further investigations that are needed to obtain a deeper insight into the mechanisms underlying the effects of SGLT2 inhibitors on the molecular, cellular, and physiological levels. Full article
(This article belongs to the Special Issue Macrophages and Inflammation)
Show Figures

Figure 1

17 pages, 803 KB  
Review
TRIM25, TRIM28 and TRIM59 and Their Protein Partners in Cancer Signaling Crosstalk: Potential Novel Therapeutic Targets for Cancer
by De Chen Chiang and Beow Keat Yap
Curr. Issues Mol. Biol. 2024, 46(10), 10745-10761; https://doi.org/10.3390/cimb46100638 - 25 Sep 2024
Cited by 3 | Viewed by 4005
Abstract
Aberrant expression of TRIM proteins has been correlated with poor prognosis and metastasis in many cancers, with many TRIM proteins acting as key oncogenic factors. TRIM proteins are actively involved in many cancer signaling pathways, such as p53, Akt, NF-κB, MAPK, TGFβ, JAK/STAT, [...] Read more.
Aberrant expression of TRIM proteins has been correlated with poor prognosis and metastasis in many cancers, with many TRIM proteins acting as key oncogenic factors. TRIM proteins are actively involved in many cancer signaling pathways, such as p53, Akt, NF-κB, MAPK, TGFβ, JAK/STAT, AMPK and Wnt/β-catenin. Therefore, this review attempts to summarize how three of the most studied TRIMs in recent years (i.e., TRIM25, TRIM28 and TRIM59) are involved directly and indirectly in the crosstalk between the signaling pathways. A brief overview of the key signaling pathways involved and their general cross talking is discussed. In addition, the direct interacting protein partners of these TRIM proteins are also highlighted in this review to give a picture of the potential protein–protein interaction that can be targeted for future discovery and for the development of novel therapeutics against cancer. This includes some examples of protein partners which have been proposed to be master switches to various cancer signaling pathways. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

13 pages, 1474 KB  
Article
Manuka Honey Inhibits Human Breast Cancer Progression in Preclinical Models
by Diana C. Márquez-Garbán, Cristian D. Yanes, Gabriela Llarena, David Elashoff, Nalo Hamilton, Mary Hardy, Madhuri Wadehra, Susan A. McCloskey and Richard J. Pietras
Nutrients 2024, 16(14), 2369; https://doi.org/10.3390/nu16142369 - 22 Jul 2024
Cited by 8 | Viewed by 34848
Abstract
Manuka honey (MH) exhibits potential antitumor activity in preclinical models of a number of human cancers. Treatment in vitro with MH at concentrations ranging from 0.3 to 5.0% (w/v) led to significant dose-dependent inhibition of proliferation of human breast [...] Read more.
Manuka honey (MH) exhibits potential antitumor activity in preclinical models of a number of human cancers. Treatment in vitro with MH at concentrations ranging from 0.3 to 5.0% (w/v) led to significant dose-dependent inhibition of proliferation of human breast cancer MCF-7 cells, but anti-proliferative effects of MH were less pronounced in MDA-MB-231 breast cancer cells. Effects of MH were also tested on non-malignant human mammary epithelial cells (HMECs) at 2.5% w/v, and it was found that MH reduced the proliferation of MCF-7 cells but not that of HMECs. Notably, the antitumor activity of MH was in the range of that exerted by treatment of MCF-7 cells with the antiestrogen tamoxifen. Further, MH treatment stimulated apoptosis of MCF-7 cells in vitro, with most cells exhibiting acute and significant levels of apoptosis that correlated with PARP activation. Additionally, the effects of MH induced the activation of AMPK and inhibition of AKT/mTOR downstream signaling. Treatment of MCF7 cells with increased concentrations of MH induced AMPK phosphorylation in a dose-dependent manner that was accompanied by inhibition of phosphorylation of AKT and mTOR downstream effector protein S6. In addition, MH reduced phosphorylated STAT3 levels in vitro, which may correlate with MH and AMPK-mediated anti-inflammatory properties. Further, in vivo, MH administered alone significantly inhibited the growth of established MCF-7 tumors in nude mice by 84%, resulting in an observable reduction in tumor volume. Our findings highlight the need for further research into the use of natural compounds, such as MH, for antitumor efficacy and potential chemoprevention and investigation of molecular pathways underlying these actions. Full article
(This article belongs to the Section Phytochemicals and Human Health)
Show Figures

Figure 1

45 pages, 1651 KB  
Review
Oxidative Stress: A Culprit in the Progression of Diabetic Kidney Disease
by Na Wang and Chun Zhang
Antioxidants 2024, 13(4), 455; https://doi.org/10.3390/antiox13040455 - 12 Apr 2024
Cited by 97 | Viewed by 13877
Abstract
Diabetic kidney disease (DKD) is the principal culprit behind chronic kidney disease (CKD), ultimately developing end-stage renal disease (ESRD) and necessitating costly dialysis or kidney transplantation. The limited therapeutic efficiency among individuals with DKD is a result of our finite understanding of its [...] Read more.
Diabetic kidney disease (DKD) is the principal culprit behind chronic kidney disease (CKD), ultimately developing end-stage renal disease (ESRD) and necessitating costly dialysis or kidney transplantation. The limited therapeutic efficiency among individuals with DKD is a result of our finite understanding of its pathogenesis. DKD is the result of complex interactions between various factors. Oxidative stress is a fundamental factor that can establish a link between hyperglycemia and the vascular complications frequently encountered in diabetes, particularly DKD. It is crucial to recognize the essential and integral role of oxidative stress in the development of diabetic vascular complications, particularly DKD. Hyperglycemia is the primary culprit that can trigger an upsurge in the production of reactive oxygen species (ROS), ultimately sparking oxidative stress. The main endogenous sources of ROS include mitochondrial ROS production, NADPH oxidases (Nox), uncoupled endothelial nitric oxide synthase (eNOS), xanthine oxidase (XO), cytochrome P450 (CYP450), and lipoxygenase. Under persistent high glucose levels, immune cells, the complement system, advanced glycation end products (AGEs), protein kinase C (PKC), polyol pathway, and the hexosamine pathway are activated. Consequently, the oxidant–antioxidant balance within the body is disrupted, which triggers a series of reactions in various downstream pathways, including phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), transforming growth factor beta/p38-mitogen-activated protein kinase (TGF-β/p38-MAPK), nuclear factor kappa B (NF-κB), adenosine monophosphate-activated protein kinase (AMPK), and the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling. The disease might persist even if strict glucose control is achieved, which can be attributed to epigenetic modifications. The treatment of DKD remains an unresolved issue. Therefore, reducing ROS is an intriguing therapeutic target. The clinical trials have shown that bardoxolone methyl, a nuclear factor erythroid 2-related factor 2 (Nrf2) activator, blood glucose-lowering drugs, such as sodium-glucose cotransporter 2 inhibitors, and glucagon-like peptide-1 receptor agonists can effectively slow down the progression of DKD by reducing oxidative stress. Other antioxidants, including vitamins, lipoic acid, Nox inhibitors, epigenetic regulators, and complement inhibitors, present a promising therapeutic option for the treatment of DKD. In this review, we conduct a thorough assessment of both preclinical studies and current findings from clinical studies that focus on targeted interventions aimed at manipulating these pathways. We aim to provide a comprehensive overview of the current state of research in this area and identify key areas for future exploration. Full article
(This article belongs to the Special Issue Oxidative Stress in Renal Health)
Show Figures

Figure 1

Back to TopTop