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

Journals

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

Search Results (252)

Search Parameters:
Keywords = immunological homeostasis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 2441 KiB  
Article
Dysfunction and Metabolic Reprogramming of Gut Regulatory T Cells in HIV-Infected Immunological Non-Responders
by Minrui Yu, Mengmeng Qu, Zerui Wang, Cheng Zhen, Baopeng Yang, Yi Zhang, Huihuang Huang, Chao Zhang, Jinwen Song, Xing Fan, Ruonan Xu, Yan-Mei Jiao and Fu-Sheng Wang
Cells 2025, 14(15), 1164; https://doi.org/10.3390/cells14151164 - 29 Jul 2025
Viewed by 359
Abstract
Disruption of the gut microenvironment is a hallmark of HIV infection, where regulatory T cells (Tregs) play a critical role in maintaining gut homeostasis. However, the mechanisms by which gut Tregs contribute to immune reconstitution failure in HIV-infected individuals remain poorly understood. In [...] Read more.
Disruption of the gut microenvironment is a hallmark of HIV infection, where regulatory T cells (Tregs) play a critical role in maintaining gut homeostasis. However, the mechanisms by which gut Tregs contribute to immune reconstitution failure in HIV-infected individuals remain poorly understood. In this study, we employed single-cell RNA sequencing (scRNA-seq) to analyze gut Treg populations across three cohorts: eight immunological responders (IRs), three immunological non-responders (INRs), and four HIV-negative controls (NCs). Our findings revealed that INRs exhibit an increased proportion of gut Tregs but with significant functional impairments, including reduced suppressive capacity and heightened apoptotic activity. Notably, these Tregs underwent metabolic reprogramming in INRs, marked by an upregulation of glycolysis-related genes and a downregulation of the oxidative phosphorylation (OXPHOS) pathway. Additionally, both the abundance of short-chain fatty acid (SCFA)-producing bacteria and SCFA concentrations were reduced in INRs. In vitro SCFA supplementation restored Treg function by enhancing suppressive capacity, reducing early apoptosis, and rebalancing cellular energy metabolism from glycolysis to OXPHOS. These findings provide a comprehensive characterization of gut Treg dysfunction in INRs and underscore the therapeutic potential of targeting gut Tregs through microbiota and metabolite supplementation to improve immune reconstitution in HIV-infected individuals. Full article
(This article belongs to the Special Issue Immune Response in HIV Infection, Pathogenesis and Persistence)
Show Figures

Graphical abstract

33 pages, 1138 KiB  
Review
Immunosenescence and the Geriatric Giants: Molecular Insights into Aging and Healthspan
by Deasy Fetarayani, Mega Kahdina, Alief Waitupu, Laras Pratiwi, Mukti Citra Ningtyas, Galih Januar Adytia and Henry Sutanto
Med. Sci. 2025, 13(3), 100; https://doi.org/10.3390/medsci13030100 - 28 Jul 2025
Viewed by 593
Abstract
Aging is associated with complex immune dysfunction that contributes to the onset and progression of the “geriatric giants”, including frailty, sarcopenia, cognitive decline, falls, and incontinence. Central to these conditions is immunosenescence, marked by thymic involution, the loss of naïve T cells, T-cell [...] Read more.
Aging is associated with complex immune dysfunction that contributes to the onset and progression of the “geriatric giants”, including frailty, sarcopenia, cognitive decline, falls, and incontinence. Central to these conditions is immunosenescence, marked by thymic involution, the loss of naïve T cells, T-cell exhaustion, impaired B-cell class switch recombination, and increased autoreactivity. Concurrently, innate immunity deteriorates due to macrophage, neutrophil, and NK cell dysfunction, while chronic low-grade inflammation—or “inflammaging”—amplifies systemic decline. Key molecular pathways such as NF-κB, mTOR, and the NLRP3 inflammasome mediate immune aging, interacting with oxidative stress, mitochondrial dysfunction, and epigenetic modifications. These processes not only impair infection control and vaccine responsiveness but also promote tissue degeneration and multimorbidity. This review explores emerging interventions—ranging from senolytics and immunonutrition to microbiome-targeted therapies and exercise—that may restore immune homeostasis and extend healthspan. Despite advances, challenges remain in translating immunological insights into clinical strategies tailored to older adults. Standardization in microbiome trials and safety optimization in senolytic therapies are critical next steps. Integrating geroscience into clinical care could help to mitigate the burden of aging-related diseases by targeting fundamental drivers of immune dysfunction. Full article
(This article belongs to the Section Immunology and Infectious Diseases)
Show Figures

Figure 1

17 pages, 2909 KiB  
Article
T Cell Dynamics in COVID-19, Long COVID and Successful Recovery
by Zoia R. Korobova, Natalia A. Arsentieva, Anastasia A. Butenko, Igor V. Kudryavtsev, Artem A. Rubinstein, Anastasia S. Turenko, Yulia V. Ostankova, Ekaterina V. Boeva, Anastasia A. Knizhnikova, Anna O. Norka, Vadim V. Rassokhin, Nikolay A. Belyakov and Areg A. Totolian
Int. J. Mol. Sci. 2025, 26(15), 7258; https://doi.org/10.3390/ijms26157258 - 27 Jul 2025
Viewed by 1619
Abstract
Despite targeting mainly the respiratory tract, SARS-CoV-2 disrupts T cell homeostasis in ways that may explain both acute lethality and long-term immunological consequences. In this study, we aimed to evaluate the T-cell-mediated chain of immunity and formation of TCR via TREC assessment in [...] Read more.
Despite targeting mainly the respiratory tract, SARS-CoV-2 disrupts T cell homeostasis in ways that may explain both acute lethality and long-term immunological consequences. In this study, we aimed to evaluate the T-cell-mediated chain of immunity and formation of TCR via TREC assessment in COVID-19 and long COVID (LC). For this study, we collected 231 blood samples taken from patients with acute COVID-19 (n = 71), convalescents (n = 51), people diagnosed with LC (n = 63), and healthy volunteers (n = 46). With flow cytometry, we assessed levels of CD4+ and CD8+ minor T cell subpopulations (i.e., naïve, central and effector memory cells (CM and EM), Th1, Th2, Th17, Tfh, Tc1, Tc2, Tc17, Tc17.1, and subpopulations of effector cells (pE1, pE2, effector cells)). Additionally, we measured TREC levels. We found distinct changes in immune cell distribution—whilst distribution of major subpopulations of T cells was similar between cohorts, we noted that COVID-19 was associated with a decrease in naïve Th and CTLs, an increase in Th2/Tc2 lymphocyte polarization, an increase in CM cells, and a decrease in effector memory cells 1,3, and TEMRA cells. LC was associated with naïve CTL increase, polarization towards Th2 population, and a decrease in Tc1, Tc2, Em2, 3, 4 cells. We also noted TREC correlating with naïve cells subpopulations. Our findings suggest ongoing immune dysregulation, possibly driven by persistent antigen exposure or tissue migration of effector cells. The positive correlation between TREC levels and naïve T cells in LC patients points to residual thymic activity. The observed Th2/Th17 bias supports the hypothesis that LC involves autoimmune mechanisms, potentially driven by molecular mimicry or loss of immune tolerance. Full article
(This article belongs to the Special Issue Long-COVID and Its Complications)
Show Figures

Figure 1

17 pages, 7852 KiB  
Article
Integrated Transcriptome and Microbiome Analyses Reveal Growth- and Stress-Response-Related Genes and Microbes in Mandarin Fish (Siniperca chuatsi)
by Fan Zhou, Wei Liu, Ming Qi, Qianrong Liang, Gaohua Yao, Cheng Ma, Xueyan Ding, Zaihang Yu, Xinyu Li and Zhanqi Wang
Fishes 2025, 10(7), 341; https://doi.org/10.3390/fishes10070341 - 10 Jul 2025
Viewed by 358
Abstract
Mandarin fish (Siniperca chuatsi) are known to exhibit distinct physiological and immunological adaptations to environmental stressors, but the underlying molecular and microbial mechanisms remain unclear. In this study, we integrated transcriptome and microbiome analyses to investigate adaptations across three geographically distinct [...] Read more.
Mandarin fish (Siniperca chuatsi) are known to exhibit distinct physiological and immunological adaptations to environmental stressors, but the underlying molecular and microbial mechanisms remain unclear. In this study, we integrated transcriptome and microbiome analyses to investigate adaptations across three geographically distinct mandarin fish groups: Guangdong (G), Qiupu (Q), and native Taihu (T). Liver RNA sequencing revealed 5339 differentially expressed genes (DEGs) between T and G and 1531 DEGs between T and Q. Functional enrichment analysis revealed group-specific responses. Specifically, DEGs from T vs. G were linked to small-molecule metabolism and innate immunity whereas the DEGs from T vs. Q were related to immune regulation and chromatin organization. The concurrent 16S rRNA sequencing of the intestinal microbiota identified 2680 amplicon sequence variants, with principal coordinate analysis showing distinct clustering (31.77% variance). Group T had higher Firmicutes abundance whereas groups G and Q had a higher relative abundance of Fusobacteriota. Correlation networks revealed key microbe–gene interactions, including positive links between Lactobacillus and immune genes in group T and negative associations with Romboutsia. These findings suggest that enhanced immune homeostasis and metabolic flexibility in group T may result from coordinated host gene expression and Lactobacillus-driven microbiome modulation. We provide new insights into the mechanisms of adaptation in mandarin fish and identify potential biomarkers for enhancing aquaculture resilience. Full article
(This article belongs to the Special Issue Fish Nutrition and Immunology)
Show Figures

Figure 1

30 pages, 722 KiB  
Review
Brain and Immune System: Intercellular Communication During Homeostasis and Neuroimmunomodulation upon Dysfunction
by Volker Schirrmacher
Int. J. Mol. Sci. 2025, 26(14), 6552; https://doi.org/10.3390/ijms26146552 - 8 Jul 2025
Viewed by 729
Abstract
The review compares the principles of organization of the brain and immune system, two important organs developed over 500 million years in multicellular organisms, including humans. It summarizes the latest results from research in neurosciences and immunology concerning intercellular communication. While in the [...] Read more.
The review compares the principles of organization of the brain and immune system, two important organs developed over 500 million years in multicellular organisms, including humans. It summarizes the latest results from research in neurosciences and immunology concerning intercellular communication. While in the brain, intercellular communication is primarily based on exchange of electrical signals, this is not the case in the immune system. The question, therefore, arises as to whether nature developed two entirely different systems of organization. It will be demonstrated that a few basic principles of brain and immune responses are organized in a different way. A majority of intercellular communications, however, such as the formation of synapses, are shown to have many similarities. Both systems are intimately interconnected to protect the body from the1 dangers of the outside and the inside world. During homeostasis, all systems are in regulatory balance. A new hypothesis states that the central systems surrounded by bone, namely the central nervous system (CNS) and the central immune system (CIS), are based on three types of stem cells and function in an open but autonomous way. T cell immune responses to antigens from blood and cerebrospinal fluid protect the system and maintain neuroimmune homeostasis. The newly discovered tunneling nanotubes and extracellular vesicles are postulated to play an important role in crosstalk with already known homeostasis regulators and help in cellular repair and the recycling of biologic material. Three examples are selected to illustrate dysfunctions of homeostasis, namely migraine, multiple sclerosis, and brain cancer. The focus on these different conditions provides deep insights into such neurological and/or immunological malfunctions. Technological advances in neurosciences and immunology can enable neuroimmunomodulation and the development of new treatment possibilities. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

26 pages, 2691 KiB  
Review
The Ovary–Liver Axis: Molecular Science and Epidemiology
by Ralf Weiskirchen and Amedeo Lonardo
Int. J. Mol. Sci. 2025, 26(13), 6382; https://doi.org/10.3390/ijms26136382 - 2 Jul 2025
Viewed by 530
Abstract
In women, gonadal hormones play a crucial regulatory role in body fat distribution and glucose–lipidic homeostasis, which are closely associated with the hepatic steatogenesis and intrahepatic inflammatory pathways. Accumulating evidence supports the idea that hepatic health is closely linked to endocrine ovarian function [...] Read more.
In women, gonadal hormones play a crucial regulatory role in body fat distribution and glucose–lipidic homeostasis, which are closely associated with the hepatic steatogenesis and intrahepatic inflammatory pathways. Accumulating evidence supports the idea that hepatic health is closely linked to endocrine ovarian function through hormonal, metabolic, and immunological communications, collectively known as the “ovary–liver axis”. This review presents the molecular mechanisms involved in sex hormone synthesis, metabolism, and signaling pathways along the ovary–liver axis, focusing on dysregulated mechanisms that may contribute to common disorders and, specifically to hepatic derangements in the context of altered ovarian function. Additionally, we analyzed epidemiological evidence supporting the ovary–liver axis, specifically examining meta-analytic studies exploring the connection between polycystic ovary syndrome and metabolic dysfunction-associated steatotic liver disease (MASLD). We also discuss studies linking hypogonadism with liver health, with a specific focus on Turner syndrome and MASLD. Furthermore, we explore the impact of menopause on liver health. Our integrated molecular and epidemiological approach identifies important clinical and public health implications, aiming to uncover potentially innovative interventions and effective strategies for managing disease progression. However, unexplored areas within the ovary–liver axis highlight the need for further research on causal pathways. Full article
(This article belongs to the Special Issue Reproductive Endocrinology Research)
Show Figures

Figure 1

22 pages, 2174 KiB  
Review
The Role of Autophagy in HIV Infection and Immunological Recovery of ART-Treated PLWH
by Mayara Sabino Leite de Oliveira Duarte, Wlisses Henrique Veloso de Carvalho-Silva and Rafael Lima Guimarães
Viruses 2025, 17(7), 884; https://doi.org/10.3390/v17070884 - 23 Jun 2025
Viewed by 613
Abstract
Human immunodeficiency virus (HIV) is responsible for acquired immunodeficiency syndrome (AIDS), a condition characterized by the depletion of CD4+ T lymphocytes, which predisposes individuals to opportunistic infections and, ultimately, death. Although antiretroviral therapy (ART) has substantially improved clinical outcomes, certain limitations persist. Notably, [...] Read more.
Human immunodeficiency virus (HIV) is responsible for acquired immunodeficiency syndrome (AIDS), a condition characterized by the depletion of CD4+ T lymphocytes, which predisposes individuals to opportunistic infections and, ultimately, death. Although antiretroviral therapy (ART) has substantially improved clinical outcomes, certain limitations persist. Notably, 15–30% of individuals undergoing ART achieve viral suppression but fail to restore adequate CD4+ T cell counts, being defined as immunological non-responders (INR) and remaining at increased risk of disease progression to AIDS. The impaired immune recovery in INRs is attributed to insufficient production and/or excessive destruction of CD4+ T lymphocytes, which can be modulated by autophagy process. This evolutionarily conserved mechanism is fundamental to lymphocyte development and activation as well as to programmed cell death pathways such as apoptosis, necroptosis, ferroptosis, and pyroptosis. These pathways are essential for understanding the impaired immune reconstitution observed in people living with HIV, whose inability to maintain immune homeostasis contributes to accelerated disease progression. This review explores the interplay between autophagy, HIV, and cell death mechanisms, highlighting its relevance in immunological recovery under ART and its potential as a therapeutic target. Full article
Show Figures

Figure 1

23 pages, 900 KiB  
Review
Perspectives on the Parathyroid–Thymus Interconnection—A Literature Review
by Maria-Paula Comănescu, Otilia Boișteanu, Delia Hînganu, Marius Valeriu Hînganu, Roxana Grigorovici and Alexandru Grigorovici
Int. J. Mol. Sci. 2025, 26(13), 6000; https://doi.org/10.3390/ijms26136000 - 23 Jun 2025
Viewed by 383
Abstract
The parathyroid and thymus glands are key components of the endocrine and immune systems, respectively, with intriguing developmental, anatomical, and functional interrelationships. This study starts from the hypothesis that, given their shared embryological origin, it is plausible that the thymus and parathyroid glands [...] Read more.
The parathyroid and thymus glands are key components of the endocrine and immune systems, respectively, with intriguing developmental, anatomical, and functional interrelationships. This study starts from the hypothesis that, given their shared embryological origin, it is plausible that the thymus and parathyroid glands interact functionally and may share pathological pathways. The present study explores the developmental pathways, spatial proximity, and potential cross-talk between these glands. Recent studies suggest that parathyroid hormone (PTH) may influence thymic function, including T-cell maturation and immune regulation, while thymic signaling molecules could impact calcium homeostasis and parathyroid activity. Understanding the functional and etiopathogenical relations between these endocrine glands offers new insights into endocrine–immunological crosstalk, and therapeutic approaches targeting disorders such as hypoparathyroidism, thymomas, myasthenia gravis and thymic hypoplasia. Perspectives and conclusion: Future research is essential to discover the molecular mechanisms underpinning this dynamic interrelation and its broader implications for health and disease. Because there is still very little data on this interaction, in-depth studies are necessary on large groups of patients. This research proposes a cross-study of the receptors for the main substances secreted by the two categories of endocrine glands. At the same time, it is essential to carry out an in-depth study on the cervico-pericardial ligaments through the lens of this glandular interaction. These ligaments could contain the main blood and nerve communication pathway between the parathyroids and the glands. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

21 pages, 1612 KiB  
Review
CD300a: An Innate Immune Checkpoint Shaping Tumor Immunity and Therapeutic Opportunity
by Jei-Ming Peng and Hui-Ying Liu
Cancers 2025, 17(11), 1786; https://doi.org/10.3390/cancers17111786 - 27 May 2025
Viewed by 1021
Abstract
CD300 family members are immunoglobulin superfamily receptors that regulate immune cell function through either activating or inhibitory signals. Among them, CD300a is a prototypical inhibitory receptor, highly expressed in both myeloid and lymphoid lineages, and plays a pivotal role in the pathogenesis of [...] Read more.
CD300 family members are immunoglobulin superfamily receptors that regulate immune cell function through either activating or inhibitory signals. Among them, CD300a is a prototypical inhibitory receptor, highly expressed in both myeloid and lymphoid lineages, and plays a pivotal role in the pathogenesis of inflammation and tumor immunity. CD300a transduces inhibitory signals in several immune cells—including mast cells, eosinophils, monocytes, dendritic cells (DCs), neutrophils, and natural killer (NK) cells—by recruiting SHP-1 phosphatase to immunoreceptor tyrosine-based inhibitory motifs (ITIMs) and suppressing activation pathways such as Toll-like receptor (TLR)-MyD88 and FcεRI signaling. Recent studies suggest that tumor cells may hijack CD300a-associated pathways to establish an immunosuppressive microenvironment that facilitates immune evasion, tumor survival, and potentially metastatic spread. Proposed mechanisms include reduced DC-mediated type I interferon (IFN) production, diminished NK cell cytotoxicity, and negative regulation of mast cell– and eosinophil-dependent anti-tumor responses. Although some of these findings are derived from in vivo models, the cumulative evidence positions CD300a as a critical immune checkpoint in tumor-associated immune regulation. In addition to its established roles in hematologic malignancies—including chronic lymphocytic leukemia, acute lymphoblastic leukemia, and acute myeloid leukemia—CD300a has also been implicated in modulating tumor-associated immune responses in other pathological contexts. While most studies emphasize its immune cell–mediated effects, emerging evidence suggests that CD300a may directly influence tumor progression by regulating immune homeostasis, intracellular signaling, and tumor microenvironment interactions. Collectively, these findings establish CD300a as a pleiotropic immunoregulatory molecule in both hematologic and non-hematologic malignancies, underscoring the need to further explore its broader relevance and therapeutic potential in cancer immunology. Full article
Show Figures

Figure 1

16 pages, 1442 KiB  
Review
Glycosylation and Acylation: Important Regulators of Immune Cell Fate Decisions
by Han Wang, Yiying Zhang, Xu Luo, Xinxin Zheng, Guangdong Bai and Junhui Liu
Biology 2025, 14(6), 611; https://doi.org/10.3390/biology14060611 - 27 May 2025
Viewed by 779
Abstract
Dissecting the determinants of immune cell fate is a central challenge in immunology and is important for understanding cell differentiation, disease diagnosis, and therapy. Post-translational modifications (PTMs) of proteins are chemical modifications of amino acids involving the addition or removal of specific groups. [...] Read more.
Dissecting the determinants of immune cell fate is a central challenge in immunology and is important for understanding cell differentiation, disease diagnosis, and therapy. Post-translational modifications (PTMs) of proteins are chemical modifications of amino acids involving the addition or removal of specific groups. As molecular gatekeepers of immune cell fate, PTMs affect immune function mainly through the regulation of important life processes such as immune cell growth, proliferation, differentiation, activation, and apoptosis. Among PTMs, glycosylation and acylation have emerged as critical regulatory mechanisms governing immune homeostasis. Through precise structural alterations of immune-related proteins and receptors, glycosylation shapes antigen recognition, cytokine signaling, and intercellular communication, while acylation regulates signal transduction and epigenetic modifications in both innate and adaptive immunity. These modifications are involved in the regulation of a wide range of biological processes and are intricately associated with the pathogenesis of many diseases, especially immune diseases. Therefore, this paper reviews the functions and mechanisms of glycosylation and acylation in regulating innate and acquired immunity, to provide new insights into the role of PTMs in disease pathogenesis and potential targeted therapies. Full article
(This article belongs to the Section Immunology)
Show Figures

Figure 1

48 pages, 2181 KiB  
Review
Tumor-Associated Macrophages: Polarization, Immunoregulation, and Immunotherapy
by Abdullah Farhan Saeed
Cells 2025, 14(10), 741; https://doi.org/10.3390/cells14100741 - 19 May 2025
Cited by 1 | Viewed by 3304
Abstract
Tumor-associated macrophages’ (TAMs) origin, polarization, and dynamic interaction in the tumor microenvironment (TME) influence cancer development. They are essential for homeostasis, monitoring, and immune protection. Cells from bone marrow or embryonic progenitors dynamically polarize into pro- or anti-tumor M2 or M1 phenotypes based [...] Read more.
Tumor-associated macrophages’ (TAMs) origin, polarization, and dynamic interaction in the tumor microenvironment (TME) influence cancer development. They are essential for homeostasis, monitoring, and immune protection. Cells from bone marrow or embryonic progenitors dynamically polarize into pro- or anti-tumor M2 or M1 phenotypes based on cytokines and metabolic signals. Recent advances in TAM heterogeneity, polarization, characterization, immunological responses, and therapy are described here. The manuscript details TAM functions and their role in resistance to PD-1/PD-L1 blockade. Similarly, TAM-targeted approaches, such as CSF-1R inhibition or PI3Kγ-driven reprogramming, are discussed to address anti-tumor immunity suppression. Furthermore, innovative biomarkers and combination therapy may enhance TAM-centric cancer therapies. It also stresses the relevance of this distinct immune cell in human health and disease, which could impact future research and therapies. Full article
Show Figures

Graphical abstract

33 pages, 4269 KiB  
Article
Peroxisome Proliferator-Activated Receptors (PPARs) May Mediate the Neuroactive Effects of Probiotic Metabolites: An In Silico Approach
by Irving Parra, Alan Carrasco-Carballo, Victoria Palafox-Sanchez, Isabel Martínez-García, José Aguilera, José L. Góngora-Alfaro, Irma Isela Aranda-González, Yousef Tizabi and Liliana Mendieta
Int. J. Mol. Sci. 2025, 26(10), 4507; https://doi.org/10.3390/ijms26104507 - 9 May 2025
Viewed by 794
Abstract
It is well established that the gut-brain axis (GBA) is a bidirectional communication between the gut and the brain. This axis, critical in maintaining overall homeostasis, is regulated at the neuronal, endocrine, and immunological levels, all of which may be influenced by the [...] Read more.
It is well established that the gut-brain axis (GBA) is a bidirectional communication between the gut and the brain. This axis, critical in maintaining overall homeostasis, is regulated at the neuronal, endocrine, and immunological levels, all of which may be influenced by the gut microbiota (GM). Therefore, dysbiosis or disruption in the GM may have serious consequences including neuroinflammation due to overactivation of the immune system. Strategies to reestablish GM integrity via use of probiotics are being pursued as novel therapeutic intervention in a variety of central and peripheral diseases. The mechanisms leading to dysbiosis or efficacy of probiotics, however, are not fully evident. Here, we performed computational analysis on two major probiotics, namely Lactobacillus Lacticaseibacillus rhamnosus GG (formerly named Lactobacillus rhamnosus, L. rhamnosus GG) and Bifidobacterium animalis spp. lactis (B. lactis or B. animalis) to not only shed some light on their mechanism(s) of action but also to identify potential molecular targets for novel probiotics. Using the PubMed web page and BioCyc Database Collection platform we specifically analyzed proteins affected by metabolites of these bacteria. Our results indicate that peroxisome proliferator-activated receptors (PPARs), nuclear receptor proteins that are involved in regulation of inflammation are key mediators of the neuroactive effect of probiotics. Full article
Show Figures

Figure 1

25 pages, 10515 KiB  
Article
Parity-Associated Differences in the Antioxidants and Fecal Microbiota of Bactrian Camels
by Hongxi Du, Jianxiong Xu, Hongcai Zhang, Jianjun Li, Fei Wang, Huan Li, Sarula Han, Jiri Gala and Jilite Wang
Vet. Sci. 2025, 12(5), 440; https://doi.org/10.3390/vetsci12050440 - 3 May 2025
Viewed by 478
Abstract
Camels survive in deserts through unique, adapted metabolic and immunological processes that are normally lethal to other species. Antioxidants and the gastrointestinal microbiota play major roles in redox homeostasis, yet they remain unexplored in camels to date. The objectives of this study were [...] Read more.
Camels survive in deserts through unique, adapted metabolic and immunological processes that are normally lethal to other species. Antioxidants and the gastrointestinal microbiota play major roles in redox homeostasis, yet they remain unexplored in camels to date. The objectives of this study were to characterize the dynamics of milk antioxidants, serum antioxidants, and the fecal microbiome of lactating Bactrian camels with different parities. In total, 30 lactating camels were selected and categorized into the following 3 groups: 10 were assigned to the first parity group (P_1), 10 were classified into the third parity group (P_3), and 10 belonged to the fifth parity group (P_5). The antioxidant parameters of the lactating camels were determined in milk and serum. The fecal microbial community of lactating camels was assessed using 16S rRNA amplicon sequencing, and the resulting library was sequenced on an Illumina NovaSeq platform. The amount of total antioxidant capacity (T-AOC) and antioxidant activity for polypepetides (DPPH) in the third parity was the highest among the groups both in the camel milk and its serum. In the case of hydroxyl radical (OH) and SOD, the amounts were 9.62 U/mL (SEM = 0.4950) and 13.64 U/mL (SEM = 0.5144), respectively, for P_1 in the serum, which were extremely significantly greater than those of the other groups. The Shannon index was significantly different between the P_1 group and either the P_3 or P_5 group. Additionally, Simpson’s diversity index significantly differed between the P_1 group and the P_5 group. Furthermore, the number of OH in camels is positively associated with the metabolic pathway of non-oxidative pentose phosphate pathway and ANAGLYCOLYSIS-PWY. In conclusion, this study revealed that different parities were associated with distinct levels of antioxidant parameters and fecal microbial ecologies in lactating Bactrian camels, where parity affects metabolic and microbial health. Understanding these dynamics in camels could optimize their nutritional management, enhancing their welfare in challenging environments. Full article
Show Figures

Figure 1

19 pages, 347 KiB  
Review
Sex-Specific Characteristics of Perivascular Fat in Aortic Aneurysms
by Katja Heller, Panagiotis Doukas, Christian Uhl and Alexander Gombert
J. Clin. Med. 2025, 14(9), 3071; https://doi.org/10.3390/jcm14093071 - 29 Apr 2025
Viewed by 591
Abstract
Aortic aneurysms (AAs), the dilation or widening of the aorta, lead to dissection or rupture with high morbidity and mortality if untreated. AA displays gender disparities in its prevalence, progression and outcomes, with women having worse outcomes and faster aneurysm growth. However, current [...] Read more.
Aortic aneurysms (AAs), the dilation or widening of the aorta, lead to dissection or rupture with high morbidity and mortality if untreated. AA displays gender disparities in its prevalence, progression and outcomes, with women having worse outcomes and faster aneurysm growth. However, current guidelines do not address gender dimorphism, emphasizing the urgent need for personalized treatment strategies and further research. Perivascular adipose tissue (PVAT), a unique type of fat surrounding blood vessels, plays a critical role in maintaining vasomotor tone and vascular homeostasis, with dysfunction associated with chronic inflammation and vessel-wall remodeling. Indeed, PVAT dysfunction promotes the development of aortic aneurysms, with hormonal and biomechanical factors exacerbating the pathological vascular microenvironment. The sexually dimorphic characteristics of PVAT include morphological, immunological, and hormonally mediated differences. Thus, targeting PVAT-mediated mechanisms may be a promising option for the (gender-specific) therapeutic management of cardiovascular pathologies. This review examines the emerging importance of PVAT in vascular health, its potential therapeutic implications for AA, and identifies gaps in the current state of research. Full article
(This article belongs to the Section Vascular Medicine)
18 pages, 3588 KiB  
Review
FNIP1 Deficiency: Pathophysiology and Clinical Manifestations of a Rare Syndromic Primary Immunodeficiency
by Samuele Roncareggi, Brian M. Iritani and Francesco Saettini
Curr. Issues Mol. Biol. 2025, 47(4), 290; https://doi.org/10.3390/cimb47040290 - 18 Apr 2025
Viewed by 582
Abstract
Folliculin-interacting protein 1 (FNIP1) is a key regulator of cellular metabolism and immune homeostasis, integrating nutrient signaling with proteostasis. FNIP1 forms a complex with folliculin (FLCN) to regulate the mechanistic target of rapamycin complex 1 (mTORC1), functioning as a GTPase-activating protein (GAP) for [...] Read more.
Folliculin-interacting protein 1 (FNIP1) is a key regulator of cellular metabolism and immune homeostasis, integrating nutrient signaling with proteostasis. FNIP1 forms a complex with folliculin (FLCN) to regulate the mechanistic target of rapamycin complex 1 (mTORC1), functioning as a GTPase-activating protein (GAP) for RagC/D. Additionally, FNIP1 interacts with heat shock protein 90 (HSP90) and undergoes phosphorylation, glycosylation, and ubiquitination, which dynamically regulate its stability and function. Evidence from murine models suggests that FNIP1 loss disrupts immune cell development and mitochondrial homeostasis. However, FNIP1 deficiency in humans remains incompletely characterized, and its full phenotypic spectrum is likely underestimated. Notably, FNIP1-deficient patients exhibit immunological and hematological abnormalities, immune dysregulation, and metabolic perturbations, emphasizing its role in cellular adaptation to stress. Understanding the mechanistic basis of FNIP1 dysfunction in human tissues will be critical for delineating its contributions to immune and metabolic disorders and identifying targeted interventions. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Biology 2025)
Show Figures

Figure 1

Back to TopTop