-
1.
Mechanism of Action and Beneficial Effects of Probiotics in Amateur and Professional Athletes.
Nami, Y, Barghi, A, Shahgolzari, M, Salehian, M, Haghshenas, B
Food science & nutrition. 2025;(1):e4658
Abstract
Probiotics are live microorganisms that, when administered in adequate amounts, provide health benefits to the host. According to the International Society of Sports Nutrition (ISSN), probiotic supplementation can optimize the health, performance, and recovery of athletes at all stages of their careers. Recent research suggests that probiotics can improve immune system functions, reduce gastrointestinal distress, and increase gut permeability in athletes. Additionally, probiotics may provide athletes with secondary health benefits that could positively affect athletic performance through enhanced recovery from fatigue, improved immune function, and maintenance of healthy gastrointestinal tract function. The integration of some probiotic strains into athletes' diets and the consumption of multi-strain compounds may lead to an improvement in performance and can positively affect performance-related aspects such as fatigue, muscle pain, body composition, and cardiorespiratory fitness. In summary, probiotics can be beneficial for athletes at all stages of their careers, from amateur to professional. This paper reviews the progress of research on the role of probiotic supplementation in improving energy metabolism and immune system functions, reducing gastrointestinal distress, and enhancing recovery from fatigue in athletes at different levels.
-
2.
Mitigating digestive complications and neutropenia in pediatric leukemia through a Persian medicine product of whole wheat-based dietary intervention: a randomized triple-blind placebo-controlled trial.
Zohalinezhad, ME, Barkhori, S, Zekavat, OR, Namjoyan, F, Bordbar, M, Hashempur, MH
Supportive care in cancer : official journal of the Multinational Association of Supportive Care in Cancer. 2025;(2):117
Abstract
Leukemia is a prevalent cancer that severely affects children, and standard chemotherapy often leads to severe gastrointestinal symptoms and neutropenia. This study aimed to discover alternative treatments to prevent neutropenia in pediatric leukemia patients and minimize chemotherapy-related complications. This randomized, placebo-controlled trial was conducted on 52 children between the ages of 3 and 18 years who were suffering from acute leukemia and undergoing chemotherapy. The study included a case and control group. A traditional wheat bran product called "Wheat Saviq" was given to the case group with Jollab syrup, while refined wheat flour and a placebo were given to the control group. For 1 month, both groups received a daily dose. Symptoms, weight, and blood cell count were measured before and after the trial. After the intervention, the pain, constipation, and bloating scores in the intervention group were lower than in the control group. Furthermore, the intervention group significantly increased white blood cells (WBC) and red blood cells (RBC). These findings suggest that incorporating wheat bran into the diet of pediatric leukemia patients has great potential in alleviating gastrointestinal symptoms and enhancing immune function. This randomized trial showed that consuming Wheat "Saviq" and Jollab syrup effectively reduced gastrointestinal symptoms and improved certain laboratory findings in children with leukemia undergoing chemotherapy. Furthermore, the results align with traditional Persian medicine (TPM) texts and further support the potential benefits of wheat bran for digestion and immune system health. IRCT registration number: IRCT20220410054474N1. Registration date: 2022-05-24, 1401/03/03.
-
3.
Immune-Boosting and Antiviral Effects of Antioxidants in COVID-19 Pneumonia: A Therapeutic Perspective.
Sanduzzi Zamparelli, S, Sanduzzi Zamparelli, A, Bocchino, M
Life (Basel, Switzerland). 2025;(1)
Abstract
The COVID-19 pandemic caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has profoundly impacted global health, with pneumonia emerging as a major complication in severe cases. The pathogenesis of COVID-19 is marked by the overproduction of reactive oxygen species (ROS) and an excessive inflammatory response, resulting in oxidative stress and significant tissue damage, particularly in the respiratory system. Antioxidants have garnered considerable attention for their potential role in managing COVID-19 pneumonia by mitigating oxidative stress and modulating immune responses. This review provides a comprehensive overview of the literature on the use of antioxidants in hospitalized patients with mild-to-moderate COVID-19. Studies exploring antioxidants, including vitamins, trace elements, nitric oxide (NO), ozone (O3), glutathione (GSH), L-carnitine, melatonin, bromelain, N-acetylcysteine (NAC), and numerous polyphenols, have yielded promising outcomes. Through their ROS-scavenging properties, these molecules support endothelial function, reduce the thrombosis risk, and may help mitigate the effects of the cytokine storm, a key contributor to COVID-19 morbidity and mortality. Clinical evidence suggests that antioxidant supplementation may improve patient outcomes by decreasing inflammation, supporting immune cell function, and potentially shortening recovery times. Furthermore, these molecules may mitigate the symptoms of COVID-19 by exerting direct antiviral effects that inhibit the infection process and genomic replication of SARS-CoV-2 in host cells. Moreover, antioxidants may work synergistically with standard antiviral treatments to reduce viral-induced oxidative damage. By integrating findings from the literature with real-world data from our clinical experience, we gain a more profound understanding of the role of antioxidants in managing COVID-19 pneumonia. Further research combining comprehensive literature reviews with real-world data analysis is crucial to validate the efficacy of antioxidants and establish evidence-based guidelines for their use in clinical practice.
-
4.
A conformational change of C-reactive protein drives neutrophil extracellular trap formation in inflammation.
Karasu, E, Halbgebauer, R, Schütte, L, Greven, J, Bläsius, FM, Zeller, J, Winninger, O, Braig, D, Messerer, DAC, Berger, B, et al
BMC biology. 2025;(1):4
Abstract
BACKGROUND C-reactive protein (CRP) represents a routine diagnostic marker of inflammation. Dissociation of native pentameric CRP (pCRP) into the monomeric structure (mCRP) liberates proinflammatory features, presumably contributing to excessive immune cell activation via unknown molecular mechanisms. RESULTS In a multi-translational study of systemic inflammation, we found a time- and inflammation-dependent pCRP dissociation into mCRP. We were able to confirm that mCRP co-localizes with leukocytes at the site of injury after polytrauma and therefore assessed whether the CRP conformation potentiates neutrophil activation. We found mCRP-induced neutrophil-extracellular trap formation in vitro and ex vivo involving nicotinamide adenine dinucleotide phosphate oxidase activation, p38/mitogen-activated protein kinase signaling, and histone H3 citrullination. Mimicking the trauma milieu in a human ex vivo whole blood model, we found significant mCRP generation as well as NET formation, prevented by blocking pCRP conformational changes. CONCLUSIONS Our data provide novel molecular insights how CRP dissociation contributes to neutrophil activation as driver of various inflammatory disorders.
-
5.
Oligomerization of STING and Chemical Regulatory Strategies.
Chen, X, Zhuo, SH, Li, YM
Chembiochem : a European journal of chemical biology. 2025;:e202400888
Abstract
Stimulator of interferon genes (STING) plays a crucial role in innate immunity. Upon the recognition of cytosolic dsDNA, STING undergoes several structural changes, with oligomerization playing a key role in initiating a cascade of immune responses. Therefore, controlling the STING pathway by manipulating STING oligomerization is a practical strategy. This review focuses on the detailed mechanism of STING oligomerization, highlighting its decisive role. It also describes oligomerization-based strategies to regulate STING protein, such as the use of small-molecule agonists and biomacromolecules, highlighting their interaction modes and potential therapeutic applications. This knowledge may lead to the development of innovative approaches for treating cancer and immune disorders.
-
6.
Redosing with Intralymphatic GAD-Alum in the Treatment of Type 1 Diabetes: The DIAGNODE-B Pilot Trial.
Casas, R, Tompa, A, Åkesson, K, Teixeira, PF, Lindqvist, A, Ludvigsson, J
International journal of molecular sciences. 2025;(1)
Abstract
Immunotherapies aimed at preserving residual beta cell function in type 1 diabetes have been successful, although the effect has been limited, or raised safety concerns. Transient effects often observed may necessitate redosing to prolong the effect, although this is not always feasible or safe. Treatment with intralymphatic GAD-alum has been shown to be tolerable and safe in persons with type 1 diabetes and has shown significant efficacy to preserve C-peptide with associated clinical benefit in individuals with the human leukocyte antigen DR3DQ2 haplotype. To further explore the feasibility and advantages of redosing with intralymphatic GAD-alum, six participants who had previously received active treatment with intralymphatic GAD-alum and carried HLA DR3-DQ2 received one additional intralymphatic dose of 4 μg GAD-alum in the pilot trial DIAGNODE-B. The participants also received 2000 U/day vitamin D (Calciferol) supplementation for two months, starting one month prior to the GAD-alum injection. During the 12-month follow-up, residual beta cell function was estimated with Mixed-Meal Tolerance Tests, and clinical and immune responses were observed. C-peptide decreased minimally, and most patients showed stable HbA1c and IDAA1c. The mean % TIR increased while the mean daily insulin dose decreased at month 12 compared to the baseline. Redosing with GAD-alum seems to be safe and tolerable, and may prolong the disease modification elicited by the original GAD-alum treatment.
-
7.
Application of Raman Spectroscopy in Non-Invasive Analysis of the Gut Microbiota and Its Impact on Gastrointestinal Health.
Krynicka, P, Koulaouzidis, G, Skonieczna-Żydecka, K, Marlicz, W, Koulaouzidis, A
Diagnostics (Basel, Switzerland). 2025;(3)
Abstract
The gut microbiota, a complex community of microorganisms, plays a crucial role in gastrointestinal (GI) health, influencing digestion, metabolism, immune function, and the gut-brain axis. Dysbiosis, or an imbalance in microbiota composition, is associated with GI disorders, including irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and colorectal cancer (CRC). Conventional microbiota analysis methods, such as next-generation sequencing (NGS) and nuclear magnetic resonance (NMR), provide valuable insights but are often expensive, time-consuming, and destructive. Raman spectroscopy (RS) is a non-invasive, cost-effective, and highly sensitive alternative. This analytical technique relies on inelastic light scattering to generate molecular "fingerprints", enabling real-time, marker-free analysis of microbiota composition and metabolic activity. This review explores the principles, sample preparation techniques, and advancements in RS, including surface-enhanced Raman spectroscopy (SERS), for microbiota research. RS facilitates identifying microbial species, analysing key metabolites like short-chain fatty acids (SCFA), and monitoring microbiota responses to dietary and therapeutic interventions. The comparative analysis highlights RS's advantages over conventional techniques, such as the minimal sample preparation, real-time capabilities, and non-destructive nature. The integration of RS with machine learning enhances its diagnostic potential, enabling biomarker discovery and personalised treatment strategies for GI disorders. Challenges, including weak Raman signals and spectral complexity, are discussed alongside emerging solutions. As RS technology advances, mainly through portable spectrometers and AI integration, its clinical application in microbiota diagnostics and personalised medicine is poised to transform GI healthcare, bridging microbiota research with practical therapeutic strategies.
-
8.
Sleep health: an unappreciated key player in colorectal cancer.
Liu, J, Yuan, Q, Zhang, Y, Wang, X, Zhai, L, Wang, R, Zheng, C, Hong, Z
Journal of Cancer. 2025;(6):1934-1943
Abstract
Colorectal cancer (CRC) poses a significant threat to human life and health. Global cancer prevalence data in 2022 indicated that the number of new cases of CRC was about 1.92 million and the deaths were around 900,000. A variety of risk factors, including genes and environment, can induce the occurrence of CRC. Previous studies have focused on the impact of dietary patterns on the development of CRC and have ignored sleep factors. Sleep deprivation is a common problem as people's work pressure increases. Sleep disorders can lead to metabolic and immune system dysregulation in people, contributing to the development and progression of many tumors. At present, there are few reports on the relationship between sleep disorders and tumors. Therefore, the purpose of this paper is to summarize and interpret the relationship between various sleep disorders and the onset and progression of CRC. This review is the first to investigate the possible mechanisms of sleep leading to CRC from the perspectives of metabolic reprogramming, intestinal microbiota disorders, and the release of inflammatory factors. In conclusion, this study highlights the rational sleep pattern and duration, which can help inhibit the occurrence of CRC.
-
9.
Therapeutic Efficacy of Intermittent Ketogenesis in Modulating Adenosine Metabolism, Immune Response, and Seizure Severity in Refractory Temporal Lobe Epilepsy: A Pilot Human Study.
Khatami, SH, Alehossein, P, Ehtiati, S, Zarei, T, Salmani, F, Bagherzadeh, S, Razmafrooz, M, Rajabibazl, M, Halimi, A, Shahmohammadi, MR, et al
Inflammation. 2025
Abstract
Temporal lobe epilepsy (TLE) is a common neurological disorder characterized by recurrent seizures originating in the temporal lobe, often affecting patients' physical, cognitive, and social well-being. Despite the availability of antiseizure medication (ASMs), approximately 30% of TLE patients exhibit drug-resistant seizures, emphasizing the need for alternative therapeutic approaches. Ketogenic diets, known for their anticonvulsant effects, have shown promise in managing drug-resistant epilepsy. However, their demanding high-fat, low-carbohydrate regimens pose significant adherence challenges. Medium-chain triglyceride (MCT) offers a viable alternative by inducing ketosis periodically without the need for continuous dietary restrictions. This study evaluated seizure severity, biochemical markers, and immune-related factors in TLE patients. The intervention group received neuro-Capridin caprylate and caprate (n-CAP), while the control group did not. Significant findings included increased plasma ATP and adenosine levels in the treatment group, along with higher expression of ADORA1 and CD73 and reduced expression of ADK. Corresponding protein changes were observed, with increased CD73 and decreased ADK levels. Caprylate and Caprate also elevated regulatory T cells and reduced proinflammatory cytokines (TNF-α, IL-6, IL-1β). These changes were associated with significant reductions in seizure severity and frequency. Intermittent ketogenesis through the consumption of Caprylate and Caprate effectively reduced seizures and improved immune and metabolic markers in drug-resistant TLE patients. These findings highlight its potential as a complementary therapy, warranting further exploration of its long-term impact and underlying molecular mechanisms.
-
10.
Prognostic roles nutritional index in patients with resectable and advanced biliary tract cancers.
Zeng, D, Wen, NY, Wang, YQ, Cheng, NS, Li, B
World journal of gastroenterology. 2025;(6):97697
Abstract
BACKGROUND Biliary tract cancer (BTC) is a rare, aggressive malignancy with increasing incidence and poor prognosis. Identifying preoperative prognostic factors is crucial for effective risk-benefit assessments and patient stratification. The prognostic nutritional index (PNI), which reflects immune-inflammatory and nutritional status, has shown prognostic value in various cancers, but its significance in BTC remains unclear. AIM: To assess the prognostic value of the preoperative PNI in BTC patients, with a focus on overall survival (OS) and disease-free survival (DFS). METHODS Comprehensive searches were conducted in the PubMed, EMBASE, and Web of Science databases from inception to April 2024. The primary outcomes of interest focused on the associations between the preoperative PNI and the prognosis of BTC patients, specifically OS and disease-free survival (DFS). Statistical analyses were conducted via STATA 17.0 software. RESULTS Seventeen studies encompassing 4645 patients met the inclusion criteria. Meta-analysis revealed that a low PNI was significantly associated with poorer OS [hazard ratio (HR) 1.91, 95%CI: 1.59-2.29; P < 0.001] and DFS (HR 1.93, 95%CI: 1.39-2.67; P < 0.001). Subgroup analyses revealed consistent results across BTC subtypes (cholangiocarcinoma and gallbladder cancer) and stages (resectable and advanced). Sensitivity analyses confirmed the robustness of these findings, and no significant publication bias was detected. CONCLUSION This study demonstrated that a low preoperative PNI predicts poor OS and DFS in BTC patients, highlighting its potential as a valuable prognostic tool. Further prospective studies are needed to validate these findings and enhance BTC patient management.