Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (8,231)

Search Parameters:
Keywords = metabolic pathways and functions

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 3725 KB  
Article
RXR Agonist V-125 Induces Distinct Transcriptional and Immunomodulatory Programs in Mammary Tumors of MMTV-Neu Mice Compared to Bexarotene
by Afrin Sultana Chowdhury, Lyndsey A. Reich, Karen T. Liby, Elizabeth S. Yeh and Ana S. Leal
Biomedicines 2026, 14(1), 80; https://doi.org/10.3390/biomedicines14010080 (registering DOI) - 30 Dec 2025
Abstract
Background: The retinoid X receptor (RXR) is a ligand-activated nuclear receptor that heterodimerizes with numerous partners to regulate diverse transcriptional programs. RXR agonists, including the FDA-approved drug bexarotene, show anti-tumor activity but are limited by adverse side effects. V-125 is a next-generation RXR [...] Read more.
Background: The retinoid X receptor (RXR) is a ligand-activated nuclear receptor that heterodimerizes with numerous partners to regulate diverse transcriptional programs. RXR agonists, including the FDA-approved drug bexarotene, show anti-tumor activity but are limited by adverse side effects. V-125 is a next-generation RXR agonist engineered for improved selectivity, pharmacokinetics, and reduced lipogenic effects. This study compares the molecular and functional effects of V-125 and bexarotene in HER2+ breast cancer models. Methods: Female MMTV-Neu mice bearing mammary tumors were treated with control, V-125 (100 mg/kg diet), or bexarotene (100 mg/kg diet) for 10 days. RNA sequencing was used to identify differentially expressed genes and pathways. Candidate targets were validated by qPCR and immunohistochemistry (IHC). Immune modulation was evaluated by IHC staining for CD8 cells and CD206+ macrophages in tumors to capture the tumor microenvironment. Functional assays in JIMT-1 human HER2+ cells assessed RXR target activation and clonogenic potential in tumor cells. Results: V-125 induced broader transcriptional changes than bexarotene, including selective upregulation of Nrg1, Nfasc, Lrrc26, and Chi3l1 genes associated with improved patient survival. Pathway analysis revealed regulation of immune activation, cancer signaling, and lipid metabolism. Both V-125 and bexarotene suppressed colony formation in JIMT-1 cells, confirming previous observations about RXR-dependent inhibition of tumor cell growth. Moreover, V-125 in vivo had distinct capabilities to increase CD8 cell infiltration and reduced CD206+ macrophages, whereas bexarotene did not. Conclusions: V-125 but not bexarotene reprograms tumor transcriptional programs and the immune landscape in an anti-tumor manner in the MMTV-neu mouse model and in in vitro models of HER2+ breast cancer. This highlights its promise as a selective RXR agonist with anti-tumor and immunomodulatory activity in HER2+ breast cancer. Full article
(This article belongs to the Special Issue Breast Cancer: New Diagnostic and Therapeutic Approaches)
Show Figures

Graphical abstract

18 pages, 1357 KB  
Review
Microplastics, Endocrine Disruptors, and Oxidative Stress: Mechanisms and Health Implications
by Kalman Kovacs, Jozsef Bodis and Reka A. A. Vass
Int. J. Mol. Sci. 2026, 27(1), 399; https://doi.org/10.3390/ijms27010399 (registering DOI) - 30 Dec 2025
Abstract
Microplastics and nanoplastics (<5 mm and <1 μm, respectively) are emerging contaminants now ubiquitous across environmental matrices and increasingly recognized for their impacts on human health. These particles commonly adsorb or contain endocrine-disrupting chemicals—such as bisphenol-A and phthalate additives—that together trigger complex biological [...] Read more.
Microplastics and nanoplastics (<5 mm and <1 μm, respectively) are emerging contaminants now ubiquitous across environmental matrices and increasingly recognized for their impacts on human health. These particles commonly adsorb or contain endocrine-disrupting chemicals—such as bisphenol-A and phthalate additives—that together trigger complex biological responses. This review examines the central role of oxidative stress in mediating the toxicity of microplastics and associated endocrine disruptors across multiple organ systems. We discuss mechanisms including cellular uptake, reactive oxygen species generation, mitochondrial dysfunction, impairment of antioxidant defenses, and activation of key signaling pathways. Organ-specific effects on reproductive health, cardiovascular function, hepatic metabolism, gut barrier integrity, and neurological systems are highlighted. Current evidence strongly supports oxidative stress as a pivotal mechanism linking microplastic exposure to systemic toxicity, underscoring important implications for public health policy and clinical intervention strategies. Full article
17 pages, 9451 KB  
Article
Broccoli Extract (Broccoli NMN®) Improves Skin Hydration by Regulating HAS and NF-κB Pathways and Reduces Wrinkle Formation via the TGF-βR1/Smad3/Collagen Pathway
by Wonhee Cho, Yeonhwa Lee, Minhee Lee, Jeongjin Park, Yuki Mukai, Dae Soo Lim, Hyelin Jeon and Woojin Jun
Curr. Issues Mol. Biol. 2026, 48(1), 50; https://doi.org/10.3390/cimb48010050 (registering DOI) - 30 Dec 2025
Abstract
This study investigated the effects and mechanisms of broccoli extract containing more than 99.0% β-NMN (BRC) on UVB-induced skin damage, including moisture loss, oxidative stress, inflammation, wrinkle formation, and melanin production, using in vitro and in vivo models. BRC treatment significantly alleviated UVB-induced [...] Read more.
This study investigated the effects and mechanisms of broccoli extract containing more than 99.0% β-NMN (BRC) on UVB-induced skin damage, including moisture loss, oxidative stress, inflammation, wrinkle formation, and melanin production, using in vitro and in vivo models. BRC treatment significantly alleviated UVB-induced skin dehydration, oxidative stress, and inflammatory responses, as well as inhibited wrinkle formation and melanin synthesis. Mechanistically, BRC enhanced skin hydration and barrier function by upregulating hyaluronic acid synthases and genes related to sphingolipid metabolism, while simultaneously suppressing NF-κB signaling and COX-2 expression, thereby re-ducing inflammation. Moreover, BRC promoted collagen synthesis by activating the TGF-βR1/Smad3/Collagen pathway and prevented extracellular matrix degradation by inhibiting JNK/c-Fos/c-Jun/MMPs signaling. In addition, BRC modulated the cAMP/PKA/CREB/MITF/TRPs pathway, leading to reduced melanin production. These findings suggest that BRC supplementation may effectively protect against UVB-induced skin damage, supporting its potential application as a functional ingredient for skin health. Full article
(This article belongs to the Section Bioorganic Chemistry and Medicinal Chemistry)
Show Figures

Figure 1

15 pages, 2283 KB  
Article
Dextrose with Insulin During Neonatal Resuscitation for Prolonged Asphyxia in a Near-Term Ovine Model: A Proof-of-Concept Study
by Praveen Chandrasekharan, Arun Prasath, Sylvia Gugino, Justin Helman, Lori Nielsen, Nicole Bradley, Mausma Bawa, Clariss Blanco, Mary Divya Kasu, Hamza Abbasi, Munmun Rawat and Jesse Slone
Children 2026, 13(1), 50; https://doi.org/10.3390/children13010050 (registering DOI) - 30 Dec 2025
Abstract
Background: Neonatal myocytes rely predominantly on glycolytic metabolism for survival during hypoxic conditions. During asphyxia, metabolic pathway dysregulation impairs cardiac myocyte contractility. Co-administration of dextrose and insulin may help restore metabolic homeostasis and improve cardiac function. Methods: Following blinded randomization and [...] Read more.
Background: Neonatal myocytes rely predominantly on glycolytic metabolism for survival during hypoxic conditions. During asphyxia, metabolic pathway dysregulation impairs cardiac myocyte contractility. Co-administration of dextrose and insulin may help restore metabolic homeostasis and improve cardiac function. Methods: Following blinded randomization and instrumentation, near-term lambs (138–140 days gestational age) were asphyxiated by umbilical cord occlusion until complete cardiac arrest, followed by 7 min of continued arrest to model severe asphyxia. Return of spontaneous circulation (ROSC) was defined as heart rate ≥ 100 beats per minute (bpm) and diastolic blood pressure ≥ 20 mmHg. Results: The incidence of ROSC was 3/6 in the control group compared to 5/5 in the experimental group receiving dextrose–insulin therapy, although this difference did not reach statistical significance. Conclusions: In this proof-of-concept study using a near-term ovine model of prolonged asphyxial cardiac arrest, dextrose and insulin co-administered with epinephrine were associated with improved ROSC rates although could be an association. Larger studies are needed to confirm these findings and evaluate clinical translation Full article
(This article belongs to the Section Pediatric Neonatology)
Show Figures

Figure 1

22 pages, 4989 KB  
Article
Immune-Modulatory Mechanism of Compound Yeast Culture in the Liver of Weaned Lambs
by Chenlu Li, Hui Bai, Pengxiang Bai, Chenxue Zhang, Yuan Wang, Dacheng Liu and Hui Chen
Animals 2026, 16(1), 104; https://doi.org/10.3390/ani16010104 (registering DOI) - 30 Dec 2025
Abstract
Compound yeast culture (CYC) is known to enhance animal health, but its effects on hepatic immune function are unclear. This study systematically examined CYC’s regulatory effects on the liver of weaned lambs using transcriptomics and integrative bioinformatics. Ten lambs were randomly assigned to [...] Read more.
Compound yeast culture (CYC) is known to enhance animal health, but its effects on hepatic immune function are unclear. This study systematically examined CYC’s regulatory effects on the liver of weaned lambs using transcriptomics and integrative bioinformatics. Ten lambs were randomly assigned to a control diet or a basal diet supplemented with 30 g/d per head of Saccharomyces cerevisiae and Kluyveromyces marxianus co-culture (CYC group) for 42 days. Histological analysis showed that CYC improved hepatocyte arrangement and sinusoidal integrity, suggesting enhanced hepatic tissue stability. Cytokine analysis revealed CYC significantly increased IL-6 and IL-1β while reducing IL-10, TGF-β1, TNF-α, and CXCL9, indicating a bidirectional modulation of the immune response. Additionally, CYC enhanced antioxidant defenses by increasing T-SOD, GSH-Px, and T-AOC activities and decreasing MDA content. Transcriptomic sequencing indicated that CYC reshaped hepatic gene expression. Upregulated genes were enriched in immune-regulatory and structural pathways, including PI3K-AKT signaling, ECM–receptor interactions, Toll-like receptor pathways, and cell adhesion molecules. Protein-level validation further confirmed activation of PI3K and AKTAKT phosphorylation with limited engagement of NF-κB signaling. Conversely, downregulated genes were mainly associated with oxidative stress and energy metabolism, such as ROS-related pathways and MAPK signaling. WGCNA identified key hub genes (PTPRC, CD86, and ITGAV), which correlate with pro-inflammatory factors and participate in immune recognition, T-cell activation, and cell adhesion. These data suggest that CYC promotes hepatic immune homeostasis by enhancing immune signaling, stabilizing tissue architecture, and modulating oxidative stress/metabolic processes. This study provides mechanistic insights into CYC’s regulation of liver immune function and supports its targeted application as a functional feed additive for ruminants. Full article
Show Figures

Figure 1

15 pages, 1080 KB  
Opinion
Intermittent Fasting and Probiotics for Gut Microbiota Modulation in Type 2 Diabetes Mellitus: A Narrative Review
by Zhiwen Zhang, Shaokang Wang, Guiju Sun and Da Pan
Nutrients 2026, 18(1), 119; https://doi.org/10.3390/nu18010119 (registering DOI) - 30 Dec 2025
Abstract
Background: Type 2 diabetes mellitus (T2DM) is a global epidemic in which gut microbiota dysbiosis contributes to impaired glucose homeostasis and chronic inflammation. Intermittent fasting (IF) and probiotic supplementation have independently demonstrated glycemic benefits in T2DM, largely through microbiota remodeling. This narrative [...] Read more.
Background: Type 2 diabetes mellitus (T2DM) is a global epidemic in which gut microbiota dysbiosis contributes to impaired glucose homeostasis and chronic inflammation. Intermittent fasting (IF) and probiotic supplementation have independently demonstrated glycemic benefits in T2DM, largely through microbiota remodeling. This narrative review synthesizes evidence up to October 2025 to clarify the microbiota-dependent mechanisms of IF and probiotics, and to evaluate the biological plausibility and preliminary clinical data for their combined application in T2DM management. Methods: We conducted a comprehensive literature review of preclinical and clinical studies (PubMed, Embase, Web of Science, and Cochrane Library) examining IF regimens (primarily time-restricted feeding and 5:2 protocols) and multi-strain probiotics containing Lactobacillus and Bifidobacterium species in T2DM or relevant models. Mechanistic pathways, microbial compositional shifts, and metabolic outcomes were qualitatively synthesized, with emphasis on overlapping signaling (short-chain fatty acids, bile acids, GLP-1, and barrier function). Results: IF consistently increases Akkermansia muciniphila and, variably, Faecalibacterium prausnitzii abundance, restores microbial circadian rhythmicity, and enhances SCFA and secondary bile acid production. Multi-strain probiotics modestly reduce HbA1c (–0.3% to –0.6%) and fasting glucose, outperforming single-strain preparations. Both interventions converge on reduced endotoxaemia and improved intestinal integrity. Preclinical models indicate potential synergy, whereas the only direct human trial to date showed neutral results. Conclusions: IF and probiotics engage overlapping microbiota-mediated pathways, supporting their combined use as an adjunctive strategy in T2DM. Adequately powered randomized trials incorporating deep metagenomics, metabolomics, and hard clinical endpoints are now required to confirm additive or synergistic efficacy. Full article
(This article belongs to the Special Issue Intermittent Fasting: Health Impacts and Therapeutic Potential)
Show Figures

Figure 1

39 pages, 2012 KB  
Review
Degenerative Disease of Intervertebral Disc: A Narrative Review of Pathogenesis, Clinical Implications and Therapies
by Lidija Gradisnik, Nina Kocivnik, Uros Maver and Tomaz Velnar
Bioengineering 2026, 13(1), 40; https://doi.org/10.3390/bioengineering13010040 (registering DOI) - 29 Dec 2025
Abstract
This narrative review examines degenerative disc disease (DDD), a major cause of chronic back pain and disability worldwide. It is a multifactorial condition resulting from a complex interplay of genetic, mechanical, metabolic, and environmental factors that progressively impair disc structure and function. The [...] Read more.
This narrative review examines degenerative disc disease (DDD), a major cause of chronic back pain and disability worldwide. It is a multifactorial condition resulting from a complex interplay of genetic, mechanical, metabolic, and environmental factors that progressively impair disc structure and function. The pathophysiology of DDD involves disruption of extracellular matrix homeostasis, cellular senescence, oxidative stress, and chronic inflammation mediated by cytokines such as IL-1β, TNF-α, and IL-6. These processes are further modulated by signalling pathways including NF-κB, MAPK, and Wnt/β-catenin, leading to matrix degradation, dehydration, and loss of disc height. Epidemiological studies highlight the contribution of lifestyle and metabolic disorders, such as obesity, smoking, and diabetes, to disease progression. Traditional conservative and surgical treatments primarily alleviate symptoms but do not halt or reverse degeneration. In contrast, recent advances in molecular biology and regenerative medicine have opened new therapeutic avenues. Mesenchymal stem cell therapy, biomaterial scaffolds, and gene-based interventions aim to restore disc homeostasis by promoting matrix synthesis and suppressing catabolic activity. Despite promising experimental results, clinical translation remains limited by challenges in cell viability, delivery methods, and long-term efficacy. Future research integrating molecular, biomechanical, and regenerative strategies offers the potential for true biological repair and disc regeneration. Full article
Show Figures

Figure 1

33 pages, 1543 KB  
Review
Biodiversity-Driven Natural Products and Bioactive Metabolites
by Giancarlo Angeles Flores, Gaia Cusumano, Roberto Venanzoni and Paola Angelini
Plants 2026, 15(1), 104; https://doi.org/10.3390/plants15010104 (registering DOI) - 29 Dec 2025
Abstract
Natural products represent one of the most diverse and functionally sophisticated groups of bioactive molecules found across plants, fungi, bacteria, and marine organisms. Recent advances in genomics, metabolomics, and chemical ecology have fundamentally redefined how these compounds are generated, regulated, and functionally deployed [...] Read more.
Natural products represent one of the most diverse and functionally sophisticated groups of bioactive molecules found across plants, fungi, bacteria, and marine organisms. Recent advances in genomics, metabolomics, and chemical ecology have fundamentally redefined how these compounds are generated, regulated, and functionally deployed in nature. Increasing evidence reveals that chemical diversity arises not solely from taxonomic lineage but from ecological pressures, evolutionary innovation, and multi-organism interactions that shape biosynthetic pathways over time. Hybrid metabolic architectures, context-dependent activation of biosynthetic gene clusters, and cross-kingdom metabolic integration collectively portray a biosynthetic landscape far more dynamic and interconnected than previously understood. At the same time, mechanistic studies demonstrate that natural products rarely act through single-target interactions. Instead, they influence redox dynamics, membrane architecture, chromatin accessibility, and intracellular signaling in distributed and synergistic ways that reflect both ecological function and evolutionary design. This review synthesizes emerging insights into the evolutionary drivers, ecological determinants, and mechanistic foundations of natural product diversity, highlighting the central role of silent biosynthetic gene clusters, meta-organismal chemistry, and network-level modes of action. By integrating these perspectives, we outline a conceptual and methodological framework capable of unlocking the vast biosynthetic potential that remains dormant within natural systems. Collectively, these advances reposition natural product research as a deeply integrative discipline at the intersection of molecular biology, ecology, evolution, and chemical innovation. Full article
Show Figures

Graphical abstract

18 pages, 2599 KB  
Article
Genomic and Metabolomic Insights Into the Probiotic Potential of Weissella viridescens
by Shuwei Zhang, Ruiting Lan, Ruiqing Zhao, Ruoshi Wang, Liyun Liu and Jianguo Xu
Biology 2026, 15(1), 63; https://doi.org/10.3390/biology15010063 (registering DOI) - 29 Dec 2025
Abstract
Weissella viridescens has been proposed as a probiotic candidate, but strain-level multi-omics evidence remains limited. The complete genome of the human-derived W. viridescens strain Wv2365 was sequenced through a hybrid assembly of Illumina and PacBio sequencing reads and compared with eight publicly available [...] Read more.
Weissella viridescens has been proposed as a probiotic candidate, but strain-level multi-omics evidence remains limited. The complete genome of the human-derived W. viridescens strain Wv2365 was sequenced through a hybrid assembly of Illumina and PacBio sequencing reads and compared with eight publicly available W. viridescens genomes. Pangenome analysis and functional annotation were performed, and metabolites were profiled by broadly targeted metabolomic analysis. In addition, the acid and bile tolerance, auto-aggregation and cell surface hydrophobicity, and antioxidant activity of the strain, as well as both in silico and phenotypic safety, were assessed. Wv2365 carries a single chromosome of 1.57 Mb with 41.3% G+C content. The species has an open pangenome with 803 core genes. Genomic and metabolomic features converged on carbohydrate and amino acid metabolism, including glycolysis/tricarboxylic acid (TCA) cycle and arginine pathways, and a carbohydrate-active enzyme (CAZyme) repertoire dominated by glycosyltransferases. In vitro, Wv2365 tolerated pH 3.0 and 0.3% bile, showed auto-aggregation, surface hydrophobicity, and 2,2-diphenyl-1-picrylhydrazyl (DPPH) and hydroxyl radical scavenging. The strain was susceptible to 10 antibiotics tested except for its intrinsic vancomycin non-susceptibility and was non-hemolytic and gelatinase negative. No acquired antimicrobial resistance or virulence genes were found in the genome. These findings indicate that W. viridescens Wv2365 is safe with probiotic traits relevant to gastrointestinal survival, colonization, and redox balance. Full article
(This article belongs to the Section Genetics and Genomics)
Show Figures

Figure 1

25 pages, 1863 KB  
Review
Autophagy–Lysosome Pathway Dysfunction in Neurodegeneration and Cancer: Mechanisms and Therapeutic Opportunities
by Mingyang Du, Yang Yu, Jiachang Wang and Cuicui Ji
Int. J. Mol. Sci. 2026, 27(1), 366; https://doi.org/10.3390/ijms27010366 (registering DOI) - 29 Dec 2025
Abstract
The autophagy–lysosome system is a master regulator of cellular homeostasis, integrating quality control, metabolism, and cell fate through the selective degradation of cytoplasmic components. Disruption of either autophagic flux or lysosomal function compromises this degradative pathway and leads to diverse pathological conditions. Emerging [...] Read more.
The autophagy–lysosome system is a master regulator of cellular homeostasis, integrating quality control, metabolism, and cell fate through the selective degradation of cytoplasmic components. Disruption of either autophagic flux or lysosomal function compromises this degradative pathway and leads to diverse pathological conditions. Emerging evidence identifies the autophagy–lysosome network as a central signaling hub that connects metabolic balance to disease progression, particularly in neurodegenerative disorders and cancer. Although cancer and neurodegenerative diseases exhibit seemingly opposite outcomes—uncontrolled proliferation versus progressive neuronal loss—both share common mechanistic foundations within the autophagy–lysosome axis. Here, we synthesize recent advances on the roles of autophagy and lysosomal mechanisms in neurodegenerative diseases and cancer, especially on how defects in lysosomal acidification, membrane integrity, and autophagosome–lysosome fusion contribute to toxic protein accumulation and organelle damage in Alzheimer’s and Parkinson’s diseases, while the same machinery is repurposed by tumor cells to sustain anabolic growth, stress tolerance, and therapy resistance. We also highlight emerging lysosome-centered therapeutic approaches, including small molecules that induce lysosomal membrane permeabilization, nanomedicine-based pH correction, and next-generation protein degradation technologies. Finally, we discuss the major challenges and future opportunities for translating these mechanistic insights into clinical interventions. Full article
(This article belongs to the Special Issue The Role of Autophagy in Disease and Cancer)
Show Figures

Figure 1

26 pages, 2869 KB  
Systematic Review
Herbal and Natural Products for Antibiotic-Associated Diarrhea: A Systematic Review of Animal Studies Focusing on Molecular Microbiome and Barrier Outcomes
by Ji Hye Hwang and You-Kyoung Choi
Pharmaceuticals 2026, 19(1), 64; https://doi.org/10.3390/ph19010064 (registering DOI) - 29 Dec 2025
Abstract
Background/Objectives: Antibiotic-associated diarrhea (AAD) arises from antibiotic-induced disruption of microbial diversity, metabolic activity, epithelial integrity, and mucosal immunity. Probiotics are widely used but often show limited efficacy under antibiotic pressure. Herbal and natural products (HNPs) may provide multi-target benefits by modulating microbiota-dependent and [...] Read more.
Background/Objectives: Antibiotic-associated diarrhea (AAD) arises from antibiotic-induced disruption of microbial diversity, metabolic activity, epithelial integrity, and mucosal immunity. Probiotics are widely used but often show limited efficacy under antibiotic pressure. Herbal and natural products (HNPs) may provide multi-target benefits by modulating microbiota-dependent and host-directed pathways. This review synthesized animal studies evaluating HNP or HNP–probiotic combination (HNP–C) therapies using molecular microbiome endpoints. Methods: Following PRISMA 2020 guidelines, controlled in vivo studies assessing HNP or HNP–C interventions for AAD were searched in Pubmed, EMBASE, Web of Science, Scopus, and CNKI through November 2025. Eligible studies reported microbial diversity, taxonomic shifts, short-chain fatty acids (SCFAs), barrier markers, or immune responses. Risk of bias was assessed using the SYRCLE tool. Due to heterogeneity, findings were narratively synthesized. Results: Twenty-seven studies met inclusion criteria (21 HNP, 6 HNP–C). HNP monotherapies consistently improved α-diversity, shifted β-diversity toward healthy controls, restored SCFA-producing taxa, and increased SCFA levels. They also enhanced tight junction proteins and reduced inflammatory cytokines. HNP–C interventions demonstrated more comprehensive microbial, epithelial, and immune recovery; however, only two studies included direct comparisons among HNP-only, probiotic-only, and combination groups. In these, HNP–C showed greater improvements than individual components, suggesting complementary or potentially complementary or additive effects. Other HNP–C studies were limited by absent comparator arms. Conclusions: HNPs appear to support recovery of microbial diversity, metabolic function, epithelial barrier integrity, and immune regulation by engaging microbiota-dependent and host-mediated mechanisms. HNP–C strategies may offer complementary benefits, although rigorously controlled comparative studies and clinical validation remain needed. Full article
(This article belongs to the Section Natural Products)
Show Figures

Graphical abstract

17 pages, 1369 KB  
Review
Coenzyme A in Brain Biology and Neurodegeneration
by Dejun Zhang, Charlie Brett, Jason Cho, Tammaryn Lashley and Ivan Gout
Biomedicines 2026, 14(1), 69; https://doi.org/10.3390/biomedicines14010069 (registering DOI) - 29 Dec 2025
Abstract
Coenzyme A (CoA) biology has been extensively studied in health and disease due to the central role of CoA in numerous metabolic and signalling processes. CoA is essential for all living organisms, and its biosynthesis and homeostasis are tightly regulated by nutrient availability, [...] Read more.
Coenzyme A (CoA) biology has been extensively studied in health and disease due to the central role of CoA in numerous metabolic and signalling processes. CoA is essential for all living organisms, and its biosynthesis and homeostasis are tightly regulated by nutrient availability, mitogenic stimuli, and stress signals. Disruptions in CoA biosynthesis, caused by inborn mutations in genes encoding enzymes of the CoA biosynthetic pathway (such as PANK2 and CoASy), lead to neurodegeneration, indicating the critical role of CoA/CoA thioesters in the function and viability of neuronal cells. The molecular mechanisms linking CoA deficiency to neurodegeneration remain unknown, but recent studies have highlighted the involvement of disrupted metabolism and redox homeostasis. The antioxidant function of CoA, mediated by protein CoAlation, has recently emerged as a novel and important mechanism of redox regulation. This review highlights well-established principles of CoA in neuronal metabolism and summarises recent advances in our understanding of its role in adaptive responses to oxidative and metabolic stress. The identification of enzymes involved in the CoAlation/deCoAlation cycle, together with the development of novel analytical tools and methodologies, may provide new insights into the discovery of more effective diagnostic and therapeutic approaches for targeting neurodegenerative diseases. Full article
Show Figures

Figure 1

25 pages, 3326 KB  
Article
Temporal Dynamics of Gene Expression and Metabolic Rewiring in Wild Barley (Hordeum spontaneum) Under Salt Stress
by Aala A. Abulfaraj and Lina Baz
Int. J. Mol. Sci. 2026, 27(1), 358; https://doi.org/10.3390/ijms27010358 (registering DOI) - 29 Dec 2025
Abstract
This study investigates the adaptive mechanisms that enable a single wild barley (Hordeum spontaneum) accession to withstand extreme salinity. Salt stress reshapes plant metabolism and gene expression, offering targets for breeding salt-tolerant cereals. A time-course RNA-Seq experiment was conducted on leaves [...] Read more.
This study investigates the adaptive mechanisms that enable a single wild barley (Hordeum spontaneum) accession to withstand extreme salinity. Salt stress reshapes plant metabolism and gene expression, offering targets for breeding salt-tolerant cereals. A time-course RNA-Seq experiment was conducted on leaves exposed to 500 mM NaCl, followed by differential expression and functional annotations to characterize transcriptomic responses. Transcriptomic profiling identified 140 dynamically upregulated genes distributed across 19 interconnected metabolic pathways, with phased activation of oxidative phosphorylation, nitrogen assimilation, lipid remodeling, and glutathione metabolism. Central metabolic nodes, including acetyl-CoA, hexadecanoyl-CoA, and ubiquinone, coordinated bioenergetic output, membrane stabilization, and redox homeostasis. Ribose-5-phosphate and ribulose-5-phosphate linked glycolysis and the pentose phosphate pathway, supplying NADPH for antioxidant defense and nucleotide repair, while riboflavin derived from Ru5P enhanced flavoprotein activity. In parallel, glucose and fructose-6-phosphate supported osmotic adjustment and glycolytic flux, and increased sterol and cuticular lipid biosynthesis, including cholesterol-like compounds, reinforced membrane integrity and calcium signaling. Glutathione and N-acetyl-glutamate together mitigated oxidative stress and modulated polyamine metabolism, strengthening cellular resilience under salt stress. These findings outline a coordinated network of metabolic and redox pathways that can guide the engineering of salt-tolerant cereals for sustainable production in saline agroecosystems. Full article
(This article belongs to the Section Molecular Plant Sciences)
Show Figures

Figure 1

14 pages, 656 KB  
Review
Cardio-Metabolic Risk in Adults Born Preterm: A Narrative Review
by Benjamim Ficial, Leonardo Gottin and Claudio Maffeis
J. Clin. Med. 2026, 15(1), 256; https://doi.org/10.3390/jcm15010256 (registering DOI) - 29 Dec 2025
Abstract
Preterm birth has evolved from being an acute neonatal challenge to a lifelong health determinant, as advances in neonatal care have markedly improved the survival of very and extremely preterm infants. This narrative review synthesizes epidemiological and mechanistic evidence linking preterm birth with [...] Read more.
Preterm birth has evolved from being an acute neonatal challenge to a lifelong health determinant, as advances in neonatal care have markedly improved the survival of very and extremely preterm infants. This narrative review synthesizes epidemiological and mechanistic evidence linking preterm birth with heightened cardiometabolic risk across the life course. In adulthood, individuals born preterm demonstrate increased rates of heart failure, ischemic heart disease, stroke, atrial fibrillation, and diabetes. Beneath these overt clinical outcomes lies a distinct phenotype characterized by increased adiposity, insulin resistance, dyslipidemia, hypertension, and atypical growth trajectories, with rapid catch-up growth amplifying long-term risk. Mechanistic pathways highlight adipose tissue maldevelopment, predisposing to metabolic syndrome, alongside cardiac maldevelopment with reduced ventricular size, impaired diastolic function, and diminished exercise capacity. Furthermore, vascular growth arrest, impaired elastin synthesis, and nephron deficiency contribute to sustained elevations in blood pressure, establishing an early substrate for hypertension and cardiovascular remodeling. These alterations reflect the developmental origins of health and disease, whereby early-life disruption of growth and maturation exerts lasting effects on organ structure and function. Collectively, the evidence identifies adults born preterm as a growing yet under-recognized patient population with a unique clinical and biochemical profile and accelerated vulnerability to non-communicable diseases. Greater awareness among pediatric and adult physicians, structured transition of care, and targeted prevention strategies are urgently needed to mitigate early cardiometabolic morbidity and optimize long-term health outcomes in this high-risk group. Full article
(This article belongs to the Special Issue New Insights in Neonatal Intensive Care)
Show Figures

Figure 1

21 pages, 1647 KB  
Review
The Donkey Genome: From Evolutionary Insights to Sustainable Breeding Strategies
by Qifei Zhu, Muhammad Zahoor Khan, Yadi Jing, Mingyang Geng, Xuemin Zhang, Yunfan Zheng, Xianggang Cao, Yongdong Peng and Changfa Wang
Animals 2026, 16(1), 93; https://doi.org/10.3390/ani16010093 (registering DOI) - 29 Dec 2025
Abstract
Donkeys (Equus asinus) are economically and ecologically important livestock species whose genetic potential remains underexplored. This review synthesizes recent advances in donkey genomics, tracing their evolutionary history while evaluating current applications in selective breeding, conservation genetics, and agricultural management. By integrating [...] Read more.
Donkeys (Equus asinus) are economically and ecologically important livestock species whose genetic potential remains underexplored. This review synthesizes recent advances in donkey genomics, tracing their evolutionary history while evaluating current applications in selective breeding, conservation genetics, and agricultural management. By integrating evidence from population genomics, functional genomics, and comparative evolutionary studies, we summarize major genomic discoveries and identify persistent knowledge gaps, with a focus on translating genomic information into practical breeding outcomes. High-quality reference genomes, population resequencing, and ancient DNA analyses have clarified the African origin, global dispersal history, and environmental adaptation of donkeys. Genome-wide approaches, including GWAS, QTL mapping, and multi-omics analyses, have further identified genes and regulatory pathways associated with thermotolerance, metabolism, reproduction, and milk production. Nevertheless, progress is still limited by small sample sizes, variable sequencing depth, and inconsistencies in phenotyping and bioinformatic pipelines, which constrain cross-population comparisons and practical applications. Addressing these challenges through standardized phenotyping, improved data integration, and collaborative research frameworks will lay the groundwork for effective conservation strategies and sustainable genomic breeding of global donkey populations. Full article
(This article belongs to the Special Issue Advances in Genetic Variability and Selection of Equines)
Show Figures

Figure 1

Back to TopTop