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

Journals

Article Types

Countries / Regions

Search Results (33)

Search Parameters:
Keywords = CCR5 disruption

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 1438 KiB  
Review
Pathogenesis of Autoimmunity/Systemic Lupus Erythematosus (SLE)
by Shunichi Shiozawa
Cells 2025, 14(14), 1080; https://doi.org/10.3390/cells14141080 - 15 Jul 2025
Viewed by 582
Abstract
SLE is characterized by the generation of a variety of autoantibodies including anti-dsDNA autoantibodies, causing damage in various organs. If autoimmunity is defined by the generation of a variety of autoantibodies against the self, SLE is the only disease to qualify. Identification of [...] Read more.
SLE is characterized by the generation of a variety of autoantibodies including anti-dsDNA autoantibodies, causing damage in various organs. If autoimmunity is defined by the generation of a variety of autoantibodies against the self, SLE is the only disease to qualify. Identification of the SLE-causing factor must fulfill the following criteria: (i) the factor induces SLE, (ii) the factor is operating in active SLE and (iii) SLE heals after removal of the factor. All candidate factors are reviewed from this viewpoint in this review. As to the cause of SLE, high levels of interferon α can induce SLE; however, interferon α in most patients did not reach this high level. BAFF (B cell activating factor of the TNF family) is increased in SLE. BAFF itself induced some manifestation of SLE, whereas removal of interferon α or BAFF by an antibody (Ab) did not heal SLE. BXSB male mice with a duplicated TLR7 gene develop SLE; however, the gene Sle1 is also required for the development of SLE. In addition, sanroque mice develop a variety of autoantibodies and SLE; the sanroque mutation, which disrupts one of the repressors of ICOS, results in increased CCR7lo CXCR5+Tfh cells, IL-21 and SLE. ICOS+T follicular helper (Tfh) cells increase in SLE and SLE-model (NZBxNZW)F1 mice, and the blockade of Tfh development ameliorated SLE, indicating the importance of Tfh cells in the pathogenesis of SLE. Self-organized criticality theory shows that SLE is caused by repeated infection, wherein SLE-inducing pathogens can vary individually depending on one’s HLA; however, the pathogen presented on HLA stimulates the T cell receptor (TCR) strongly beyond self-organized criticality. This stimulation generates TCR-revised, autoreactive DOCK8+Tfh cells, which induced a variety of autoantibodies and SLE. The SARS-CoV-2 virus is an example pathogen because SLE occurs after SARS-CoV-2 infection and vaccination. DOCK8+Tfh cells and SLE decreased after conventional or anti-DOCK Ab therapies. Thus, DOCK8+Tfh cells newly generated after repeated infection fulfill the criteria (i), (ii) and (iii) as the cause of SLE. Full article
Show Figures

Figure 1

30 pages, 878 KiB  
Article
Berth Efficiency Under Risk Conditions in Seaports Through Integrated DEA and AHP Analysis
by Deda Đelović, Marinko Aleksić, Oto Iker and Michail Chalaris
J. Mar. Sci. Eng. 2025, 13(7), 1324; https://doi.org/10.3390/jmse13071324 - 10 Jul 2025
Viewed by 312
Abstract
In the context of increasingly complex and dynamic maritime logistics, seaports serve as critical nodes for intermodal transport, energy distribution, and global trade. Ensuring the safe and uninterrupted operation of port infrastructure—particularly berths—is vital for maintaining supply chain resilience. This study explores the [...] Read more.
In the context of increasingly complex and dynamic maritime logistics, seaports serve as critical nodes for intermodal transport, energy distribution, and global trade. Ensuring the safe and uninterrupted operation of port infrastructure—particularly berths—is vital for maintaining supply chain resilience. This study explores the impact of multiple risk categories on berth efficiency in a seaport, aligning with the growing emphasis on maritime safety and risk-informed decision-making. A two-stage methodology is adopted. In the first phase, the DEA CCR input-oriented model is employed to assess the efficiency of selected berths considered as Decision Making Units (DMUs). In the second phase, the Analytical Hierarchy Process (AHP) is used to categorize and quantify the impact of four major risk classes—operational, technical, safety, and environmental—on berth efficiency. The results demonstrate that operational and safety risks contribute 63.91% of the composite weight in the AHP risk assessment hierarchy. These findings are highly relevant to contemporary efforts in maritime risk modeling, especially for individual ports and port systems with high berth utilization and vulnerability to system disruptions. The proposed integrated approach offers a scalable and replicable decision-support tool for port authorities, port operators, planners, and maritime safety stakeholders, enabling proactive risk mitigation, optimal utilization of available resources in a port, and improved berth performance. Its methodological design is appropriately suited to support further applications in port resilience frameworks and maritime safety strategies, being one of the bases for establishing collision avoidance strategies related to an individual port and/or port system, too. Full article
(This article belongs to the Special Issue Recent Advances in Maritime Safety and Ship Collision Avoidance)
Show Figures

Figure 1

17 pages, 2381 KiB  
Review
The Genetic Fingerprint of HIV in the Brain: Insights into Neurocognitive Dysfunction
by Sushama Jadhav, Shreeya Nair and Vijay Nema
Neuroglia 2025, 6(2), 23; https://doi.org/10.3390/neuroglia6020023 - 9 Jun 2025
Viewed by 729
Abstract
HIV, primarily targeting CD4 cells, infiltrates the CNS through various mechanisms, including chemokine-mediated signaling and blood–brain barrier disruption, leading to neuroinflammation and neuronal dysfunction. Viral proteins such as gp120, Tat, and Vpr directly induce neurotoxicity, oxidative stress, and mitochondrial dysfunction, exacerbating cognitive deficits [...] Read more.
HIV, primarily targeting CD4 cells, infiltrates the CNS through various mechanisms, including chemokine-mediated signaling and blood–brain barrier disruption, leading to neuroinflammation and neuronal dysfunction. Viral proteins such as gp120, Tat, and Vpr directly induce neurotoxicity, oxidative stress, and mitochondrial dysfunction, exacerbating cognitive deficits and motor impairments observed in HIV-associated neurocognitive disorders (HANDs). Host genetic factors, including CCR5 mutations and HLA alleles, influence susceptibility to HIV-related neurologic complications, shaping disease progression and treatment responses. Advanced molecular and bioinformatics techniques, from genome sequencing to structural modeling and network analysis, provide insights into viral pathogenesis and identify potential therapeutic targets. These findings underscore the future potential of precision medicine approaches tailored to individual genetic profiles to mitigate neurologic complications and improve outcomes in HIV-infected populations. This comprehensive review explores the intricate interplay between HIV infection and neurogenetics, focusing on how the virus impacts the central nervous system (CNS) and contributes to neurocognitive disorders. This report delves into how the virus influences genetic expression, neuroinflammation, and neurodegeneration, offering insights into molecular mechanisms behind HAND. Full article
Show Figures

Figure 1

18 pages, 1935 KiB  
Review
Progress in CRISPR Technology for Antiviral Treatments: Genome Editing as a Potential Cure for Chronic Viral Infections
by Fatemeh Nouri, Farnaz Alibabaei, Behina Forouzanmehr, Hamed Tahmasebi, Valentyn Oksenych and Majid Eslami
Microbiol. Res. 2025, 16(5), 104; https://doi.org/10.3390/microbiolres16050104 - 20 May 2025
Viewed by 1721
Abstract
The CRISPR–Cas system has transformed molecular biology by providing precise tools for genome editing and pathogen detection. Originating from bacterial adaptive immunity, CRISPR technology identifies and cleaves genetic material from pathogens, thereby preventing infections. CRISPR–Cas9, the most widely utilized variant, creates double-stranded breaks [...] Read more.
The CRISPR–Cas system has transformed molecular biology by providing precise tools for genome editing and pathogen detection. Originating from bacterial adaptive immunity, CRISPR technology identifies and cleaves genetic material from pathogens, thereby preventing infections. CRISPR–Cas9, the most widely utilized variant, creates double-stranded breaks in the target DNA, enabling genetic disruptions or edits. This approach has shown significant potential in antiviral therapies, addressing chronic infections, such as HIV, SARS-CoV-2, and hepatitis viruses. In HIV, CRISPR–Cas9 edits the essential viral genes and disrupts latent reservoirs, while CCR5 gene modifications render the T cells resistant to viral entry. Similarly, SARS-CoV-2 is targeted using CRISPR–Cas13d to inhibit the conserved viral genes, significantly reducing viral loads. Hepatitis B and C treatments leverage CRISPR technologies to target conserved genomic regions, limiting replication and expression. Emerging innovations, such as the PAC-MAN approach for influenza and base-editing systems to reduce off-target effects, further highlight the therapeutic versatility of CRISPR. Additionally, advances in Cas12a and Cas13 have driven the development of diagnostic platforms like DETECTR and SHERLOCK, which provide rapid and cost-effective viral detection. Innovative tools like AIOD-CRISPR enable accessible point-of-care diagnostics for early viral detection. Experimental approaches, such as targeting latent HSV-1 reservoirs, highlight the transformative potential of CRISPR in combating persistent infections. Full article
Show Figures

Figure 1

23 pages, 5381 KiB  
Article
Characterization of OsCAF1 Protein Function in Rice Response to Thermal Stress
by Vu-Bao Nguyen and Chung-An Lu
Plants 2025, 14(7), 1036; https://doi.org/10.3390/plants14071036 - 27 Mar 2025
Cited by 1 | Viewed by 778
Abstract
Heat stress is a critical environmental challenge that disrupts rice growth, development, and productivity and poses a significant threat to global food security. The CCR4-NOT protein complex, particularly its CCR4-associated factor 1 (CAF1) subunit, plays a crucial role in the dynamic regulation of [...] Read more.
Heat stress is a critical environmental challenge that disrupts rice growth, development, and productivity and poses a significant threat to global food security. The CCR4-NOT protein complex, particularly its CCR4-associated factor 1 (CAF1) subunit, plays a crucial role in the dynamic regulation of gene expression by mediating mRNA de-adenylation, a key step in mRNA degradation and turnover. However, the specific function of OsCAF1 proteins under heat stress in rice remains poorly understood. In this study, we investigated the dynamic subcellular localization of OsCAF1A in response to elevated temperatures and its role in heat stress tolerance. Under normal conditions, OsCAF1A is diffusely localized to the cytoplasm. However, OsCAF1A predominantly localizes to processing bodies (PBs) under heat stress. The results of interaction studies revealed that two DEAD-box RNA helicases, OseIF4AIIb and OsRH8, modulate the re-localization of OsCAF1A, by OseIF4AIIb inhibiting and OsRH8 promoting its association with PBs during heat stress. Furthermore, OsCAF1A mRNA was more abundantly expressed in rice seedlings than other OsCAF1 genes and is further upregulated by high temperature. The overexpression of OsCAF1A significantly enhanced heat tolerance, whereas mutants exhibited increased heat sensitivity. These findings underscore the potential of OsCAF1A as a tool to improve crop resilience to climate change. Full article
(This article belongs to the Special Issue Plant Metabolic Responses to Biotic and Abiotic Stress—2nd Edition)
Show Figures

Figure 1

24 pages, 2285 KiB  
Review
The Function of Myostatin in Ameliorating Bone Metabolism Abnormalities in Individuals with Type 2 Diabetes Mellitus by Exercise
by Chenghao Zhong, Xinyu Zeng, Xiaoyan Yi, Yuxin Yang, Jianbo Hu, Rongbin Yin and Xianghe Chen
Curr. Issues Mol. Biol. 2025, 47(3), 158; https://doi.org/10.3390/cimb47030158 - 27 Feb 2025
Cited by 2 | Viewed by 1391
Abstract
Purpose: The molecular mechanisms involved in bone metabolism abnormalities in individuals with type 2 diabetes mellitus (T2DM) are a prominent area of investigation within the life sciences field. Myostatin (MSTN), a member of the TGF-β superfamily, serves as a critical negative regulator of [...] Read more.
Purpose: The molecular mechanisms involved in bone metabolism abnormalities in individuals with type 2 diabetes mellitus (T2DM) are a prominent area of investigation within the life sciences field. Myostatin (MSTN), a member of the TGF-β superfamily, serves as a critical negative regulator of skeletal muscle growth and bone metabolism. Current research on the exercise-mediated regulation of MSTN expression predominantly focuses on its role in skeletal muscle. However, due to the intricate and multifaceted mechanical and biochemical interactions between muscle and bone, the precise mechanisms by which exercise modulates MSTN to enhance bone metabolic disorders in T2DM necessitate additional exploration. The objective of this review is to systematically synthesize and evaluate the role of MSTN in the development of bone metabolism disorders associated with T2DM and elucidate the underlying mechanisms influenced by exercise interventions, aiming to offer novel insights and theoretical recommendations for enhancing bone health through physical activity. Methods: Relevant articles in Chinese and English up to July 2024 were selected using specific search terms and databases (PubMed, CNKI, Web of Science); 147 studies were finally included after evaluation, and the reference lists were checked for other relevant research. Results: Myostatin’s heightened expression in the bone and skeletal muscle of individuals with T2DM can impede various pathways, such as PI3K/AKT/mTOR and Wnt/β-catenin, hindering osteoblast differentiation and bone mineralization. Additionally, it can stimulate osteoclast differentiation and bone resorption capacity by facilitating Smad2-dependent NFATc1 nuclear translocation and PI3K/AKT/AP-1-mediated pro-inflammatory factor expression pathways, thereby contributing to bone metabolism disorders. Physical exercise plays a crucial role in ameliorating bone metabolism abnormalities in individuals with T2DM. Exercise can activate pathways like Wnt/GSK-3β/β-catenin, thereby suppressing myostatin and downstream Smads, CCL20/CCR6, and Nox4 target gene expression, fostering bone formation, inhibiting bone resorption, and enhancing bone metabolism in T2DM. Conclusion: In the context of T2DM, MSTN has been shown to exacerbate bone metabolic disorders by inhibiting the differentiation of osteoblasts and the process of bone mineralization while simultaneously promoting the differentiation and activity of osteoclasts. Exercise interventions have demonstrated efficacy in downregulating MSTN expression, disrupting its downstream signaling pathways, and enhancing bone metabolism. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

24 pages, 3160 KiB  
Article
Inherited Dyslipidemic Splenomegaly: A Genetic Macrophage Storage Disorder Caused by Disruptive Apolipoprotein E (APOE) Variants
by Elise A. Ferreira, Machteld M. Oud, Saskia N. van der Crabben, Miranda Versloot, Susan M. I. Goorden, Clara D. M. van Karnebeek, Jeffrey Kroon and Mirjam Langeveld
Genes 2025, 16(3), 289; https://doi.org/10.3390/genes16030289 - 27 Feb 2025
Viewed by 1102
Abstract
Background: Persistent splenomegaly, often an incidental finding, can originate from a number of inherited metabolic disorders (IMDs). Variants of APOE are primarily known as risk factors in terms of cardiovascular disease; however, severe dysfunction of APOE can result in a disease phenotype with [...] Read more.
Background: Persistent splenomegaly, often an incidental finding, can originate from a number of inherited metabolic disorders (IMDs). Variants of APOE are primarily known as risk factors in terms of cardiovascular disease; however, severe dysfunction of APOE can result in a disease phenotype with considerable overlap with lysosomal storage disorders (LSDs), including splenomegaly and gross elevation of N-palmitoyl-O-phosphocholine-serine (PPCS). Methods: A case study (deep phenotyping, genetic and FACS analysis) and literature study was conducted. Results: The index patient, with a family history of early-onset cardiovascular disease, presented with splenic infarctions in a grossly enlarged spleen. The identified genetic cause was homozygosity for two APOE variants (c.604C>T, p.(Arg202Cys) and c.512G>A, p.(Gly171Asp); ε1/ε1), resulting in a macrophage storage phenotype resembling an LSD that was also present in the brother of the index patient. A FACS analysis of the circulating monocytes showed increased lipid content and the expression of activation markers (CD11b, CCR2, CD36). This activated state enhances lipoprotein intake, which eventually converts these monocytes/macrophages into foam cells, accumulating in tissues (e.g., spleen and vascular wall). A literature search identified seven individuals with splenomegaly caused by APOE variants (deletion of leucine at position 167). The combined data from all patients identified male gender, splenectomy and obesity as potential modifiers determining the severity of the phenotype (i.e., degree of triglyceride increase in plasma and/or spleen size). Symptoms are (partially) reversible by lipid-lowering medication and energy restricted diets and splenectomy is contra-indicated. Conclusions: Inherited dyslipidemic splenomegaly caused by disruptive APOE variants should be included in the differential diagnoses of unexplained splenomegaly with abnormal lipid profiles. A plasma lipid profile consistent with dysbetalipoproteinemia is a diagnostic biomarker for this IMD. Full article
(This article belongs to the Collection Genetics and Genomics of Rare Disorders)
Show Figures

Figure 1

24 pages, 10194 KiB  
Article
Multi-Omics Research Reveals the Effects of the ABA-Regulated Phenylpropanoid Biosynthesis Pathway on the UV-B Response in Rhododendron chrysanthum Pall.
by Wang Yu, Xiangru Zhou, Jinhao Meng, Xiaofu Zhou and Hongwei Xu
Plants 2025, 14(1), 101; https://doi.org/10.3390/plants14010101 - 1 Jan 2025
Cited by 3 | Viewed by 1208
Abstract
The growing depletion of the ozone layer has led to increased ultraviolet B (UV-B) radiation, prompting plants like the alpine Rhododendron chrysanthum Pall. (R. chrysanthum) to adapt to these harsh conditions. This study explored how abscisic acid (ABA) signaling influences R. [...] Read more.
The growing depletion of the ozone layer has led to increased ultraviolet B (UV-B) radiation, prompting plants like the alpine Rhododendron chrysanthum Pall. (R. chrysanthum) to adapt to these harsh conditions. This study explored how abscisic acid (ABA) signaling influences R. chrysanthum’s metabolic responses under UV-B stress. R. chrysanthum was treated with UV-B radiation and exogenous ABA for widely targeted metabolomics, transcriptomics, and proteomics assays, and relevant chlorophyll fluorescence parameters were also determined. It was observed that UV-B stress negatively impacts the plant’s photosynthetic machinery, disrupting multiple metabolic processes. Multi-omics analysis revealed that ABA application mitigates the detrimental effects of UV-B on photosynthesis and bolsters the plant’s antioxidant defenses. Additionally, both UV-B exposure and ABA treatment significantly influenced the phenylpropanoid biosynthesis pathway, activating key enzyme genes, such as 4CL, CCR, and HCT. The study also highlighted the MYB–bHLH–WD40 (MBW) complex’s role in regulating this pathway and its interaction with ABA signaling components. These findings underscore ABA’s crucial function in improving plant resistance to UV-B stress and offer novel insights into plant stress biology. Full article
(This article belongs to the Special Issue Responses of Crops to Abiotic Stress)
Show Figures

Figure 1

12 pages, 2210 KiB  
Article
Clinical and Cytogenetic Impact of Maternal Balanced Double Translocation: A Familial Case of 15q11.2 Microduplication and Microdeletion Syndromes with Genetic Counselling Implications
by Daniela Koeller R. Vieira, Ingrid Bendas Feres Lima, Carla Rosenberg, Carlos Roberto da Fonseca, Leonardo Henrique Ferreira Gomes, Letícia da Cunha Guida, Patrícia Camacho Mazzonetto, Juan Llerena and Elenice Ferreira Bastos
Genes 2024, 15(12), 1546; https://doi.org/10.3390/genes15121546 - 29 Nov 2024
Viewed by 1698
Abstract
Background: Balanced chromosomal translocations occur in approximately 0.16 to 0.20% of live births. While most carriers are phenotypically normal, they are at risk of generating unbalanced gametes during meiosis, leading to genetic anomalies such as aneuploidies, deletions, duplications, and gene disruptions. These anomalies [...] Read more.
Background: Balanced chromosomal translocations occur in approximately 0.16 to 0.20% of live births. While most carriers are phenotypically normal, they are at risk of generating unbalanced gametes during meiosis, leading to genetic anomalies such as aneuploidies, deletions, duplications, and gene disruptions. These anomalies can result in spontaneous abortions or congenital anomalies, including neurodevelopmental disorders. Complex chromosomal rearrangements (CCRs) involving more than two chromosomes are rare but further increase the probability of producing unbalanced gametes. Neurodevelopmental disorders such as Angelman syndrome (AS) and duplication 15q11q13 syndrome (Dup15q) are associated with such chromosomal abnormalities. Methods: This study describes a family with a de novo maternal balanced double translocation involving chromosomes 13, 19, and 15, resulting in two offspring with unbalanced chromosomal abnormalities. Cytogenetic evaluations were performed using GTG banding, fluorescence in situ hybridization (FISH), and low-pass whole-genome sequencing (LP-WGS). Methylation analysis was conducted using methylation-sensitive high-resolution melting (MS-HRM) to diagnose Angelman syndrome. Results: The cytogenetic and molecular analyses identified an 8.9 Mb duplication in 15q11.2q13.3 in one child, and an 8.9 Mb deletion in the same region in the second child. Both abnormalities affected critical neurodevelopmental genes, such as SNRPN. FISH and MS-HRM confirmed the chromosomal imbalances and the diagnosis of Angelman syndrome in the second child. The maternal balanced translocation was found to be cryptic, contributing to the complex inheritance pattern. Conclusion: This case highlights the importance of using multiple genetic platforms to uncover complex chromosomal rearrangements and their impact on neurodevelopmental disorders. The findings underscore the need for thorough genetic counseling, especially in families with such rare chromosomal alterations, to manage reproductive outcomes and neurodevelopmental risks. Full article
(This article belongs to the Section Genetic Diagnosis)
Show Figures

Figure 1

15 pages, 1937 KiB  
Article
Chemokinergic and Dopaminergic Signalling Collaborates through the Heteromer Formed by CCR9 and Dopamine Receptor D5 Increasing the Migratory Speed of Effector CD4+ T-Cells to Infiltrate the Colonic Mucosa
by Javier Campos, Francisco Osorio-Barrios, Felipe Villanelo, Sebastian E. Gutierrez-Maldonado, Pablo Vargas, Tomás Pérez-Acle and Rodrigo Pacheco
Int. J. Mol. Sci. 2024, 25(18), 10022; https://doi.org/10.3390/ijms251810022 - 18 Sep 2024
Cited by 3 | Viewed by 1547
Abstract
Inflammatory bowel diseases (IBDs) involve chronic inflammation of the gastrointestinal tract, where effector CD4+ T-cells play a central role. Thereby, the recruitment of T-cells into the colonic mucosa represents a key process in IBD. We recently found that CCR9 and DRD5 might [...] Read more.
Inflammatory bowel diseases (IBDs) involve chronic inflammation of the gastrointestinal tract, where effector CD4+ T-cells play a central role. Thereby, the recruitment of T-cells into the colonic mucosa represents a key process in IBD. We recently found that CCR9 and DRD5 might form a heteromeric complex on the T-cell surface. The increase in CCL25 production and the reduction in dopamine levels associated with colonic inflammation represent a dual signal stimulating the CCR9:DRD5 heteromer, which promotes the recruitment of CD4+ T-cells into the colonic lamina propria. Here, we aimed to analyse the molecular requirements involved in the heteromer assembly as well as to determine the underlying cellular mechanisms involved in the colonic tropism given by the stimulation of the CCR9:DRD5 complex. The results show that dual stimulation of the CCR9:DRD5 heteromer potentiates the phosphorylation of the myosin light chain 2 (MLC2) and the migration speed in confined microchannels. Accordingly, disrupting the CCR9:DRD5 assembly induced a sharp reduction in the pMLC2 in vitro, decreased the migratory speed in confined microchannels, and dampened the recruitment of CD4+ T-cells into the inflamed colonic mucosa. Furthermore, in silico analysis confirmed that the interface of interaction of CCR9:DRD5 is formed by the transmembrane segments 5 and 6 from each protomer. Our findings demonstrated that the CCR9:DRD5 heteromeric complex plays a fundamental role in the migration of CD4+ T-cells into the colonic mucosa upon inflammation. Thereby, the present study encourages the design of strategies for disassembling the formation of the CCR9:DRD5 as a therapeutic opportunity to treat IBD. Full article
(This article belongs to the Special Issue Inflammatory Bowel Disease: Molecular Insights)
Show Figures

Figure 1

18 pages, 3562 KiB  
Article
Transcriptome Analysis Reveals the Immunosuppression in Tiger Pufferfish (Takifugu rubripes) under Cryptocaryon irritans Infection
by Yong Chi, Robert Mukiibi, Hongxiang Zhang, Haien Zhang, Weidong Li, Diego Robledo, Songlin Chen and Yangzhen Li
Animals 2024, 14(14), 2058; https://doi.org/10.3390/ani14142058 - 13 Jul 2024
Viewed by 1806
Abstract
The tiger pufferfish (Takifugu rubripes), also known as fugu, has recently suffered from severe C. irritans infections under aquaculture environment, yet the underlying immune mechanisms against the parasite remain poorly understood. In this study, we conducted a comprehensive transcriptome analysis of [...] Read more.
The tiger pufferfish (Takifugu rubripes), also known as fugu, has recently suffered from severe C. irritans infections under aquaculture environment, yet the underlying immune mechanisms against the parasite remain poorly understood. In this study, we conducted a comprehensive transcriptome analysis of the gill tissue from infected and uninfected fish using PacBio long-read (one pooled sample each for seriously infected and healthy individuals, respectively) and Illumina short-read (three pools for mildly infected, seriously infected, and healthy individuals, respectively) RNA sequencing technologies. After aligning sequence data to fugu’s reference genome, 47,307 and 34,413 known full-length transcripts were identified and profiled in healthy and infected fish, respectively. Similarly, we identified and profiled 1126 and 803 novel genes that were obtained from healthy and infected fish, respectively. Interestingly, we found a decrease in the number of alternative splicing (AS) events and long non-coding RNAs (lncRNAs) after infection with C. irritans, suggesting that they may be involved in the regulation of the immune response in fugu. There were 687 and 1535 differentially expressed genes (DEGs) in moderately and heavily infected fish, respectively, compared to uninfected fish. Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed that immune-related DEGs in the two comparison groups were mainly enriched in cytokine-cytokine receptor interactions, ECM-receptor interactions, T-cell receptor signaling pathways, Th1 and Th2 cell differentiation, and Th17 cell differentiation pathways. Further analysis revealed that a large number of immune-related genes were downregulated in infected fish relative to uninfected ones, such as CCR7, IL7R, TNFRSF21, CD4, COL2A1, FOXP3B, and ITGA8. Our study suggests that C. irritans is potentially a highly efficient parasite that may disrupt the defense mechanisms of fugu against it. In addition, in combination of short-read RNA sequencing and previous genome-wide association analyses, we identified five key genes (NDUFB6, PRELID1, SMOX, SLC25A4, and DENND1B) that might be closely associated with C. irritans resistance. This study not only provides valuable resources of novel genic transcripts for further research, but also provides new insights into the immune mechanisms underlying C. irritans infection response in farmed fugu. Full article
Show Figures

Figure 1

14 pages, 3276 KiB  
Article
Nicotinamide Riboside Augments Human Macrophage Migration via SIRT3-Mediated Prostaglandin E2 Signaling
by Jing Wu, Maximilian Bley, Russell S. Steans, Allison M. Meadows, Rebecca D. Huffstutler, Rong Tian, Julian L. Griffin and Michael N. Sack
Cells 2024, 13(5), 455; https://doi.org/10.3390/cells13050455 - 5 Mar 2024
Cited by 1 | Viewed by 3366
Abstract
NAD+ boosting via nicotinamide riboside (NR) confers anti-inflammatory effects. However, its underlying mechanisms and therapeutic potential remain incompletely defined. Here, we showed that NR increased the expression of CC-chemokine receptor 7 (CCR7) in human M1 macrophages by flow cytometric analysis of cell [...] Read more.
NAD+ boosting via nicotinamide riboside (NR) confers anti-inflammatory effects. However, its underlying mechanisms and therapeutic potential remain incompletely defined. Here, we showed that NR increased the expression of CC-chemokine receptor 7 (CCR7) in human M1 macrophages by flow cytometric analysis of cell surface receptors. Consequently, chemokine ligand 19 (CCL19, ligand for CCR7)-induced macrophage migration was enhanced following NR administration. Metabolomics analysis revealed that prostaglandin E2 (PGE2) was increased by NR in human monocytes and in human serum following in vivo NR supplementation. Furthermore, NR-mediated upregulation of macrophage migration through CCL19/CCR7 was dependent on PGE2 synthesis. We also demonstrated that NR upregulated PGE2 synthesis through SIRT3-dependent post-transcriptional regulation of cyclooxygenase 2 (COX-2). The NR/SIRT3/migration axis was further validated using the scratch-test model where NR and SIRT3 promoted more robust migration across a uniformly disrupted macrophage monolayer. Thus, NR-mediated metabolic regulation of macrophage migration and wound healing may have therapeutic potential for the topical management of chronic wound healing. Full article
(This article belongs to the Special Issue Advances in Leukocyte Migration and Location in Health and Disease)
Show Figures

Figure 1

25 pages, 4204 KiB  
Article
Chemokine Receptor Antagonists Prevent and Reverse Cofilin-Actin Rod Pathology and Protect Synapses in Cultured Rodent and Human iPSC-Derived Neurons
by Thomas B. Kuhn, Laurie S. Minamide, Lubna H. Tahtamouni, Sydney A. Alderfer, Keifer P. Walsh, Alisa E. Shaw, Omar Yanouri, Henry J. Haigler, Michael R. Ruff and James R. Bamburg
Biomedicines 2024, 12(1), 93; https://doi.org/10.3390/biomedicines12010093 - 1 Jan 2024
Cited by 2 | Viewed by 4202
Abstract
Synapse loss is the principal cause of cognitive decline in Alzheimer’s disease (AD) and related disorders (ADRD). Synapse development depends on the intricate dynamics of the neuronal cytoskeleton. Cofilin, the major protein regulating actin dynamics, can be sequestered into cofilactin rods, intra-neurite bundles [...] Read more.
Synapse loss is the principal cause of cognitive decline in Alzheimer’s disease (AD) and related disorders (ADRD). Synapse development depends on the intricate dynamics of the neuronal cytoskeleton. Cofilin, the major protein regulating actin dynamics, can be sequestered into cofilactin rods, intra-neurite bundles of cofilin-saturated actin filaments that can disrupt vesicular trafficking and cause synaptic loss. Rods are a brain pathology in human AD and mouse models of AD and ADRD. Eliminating rods is the focus of this paper. One pathway for rod formation is triggered in ~20% of rodent hippocampal neurons by disease-related factors (e.g., soluble oligomers of Amyloid-β (Aβ)) and requires cellular prion protein (PrPC), active NADPH oxidase (NOX), and cytokine/chemokine receptors (CCRs). FDA-approved antagonists of CXCR4 and CCR5 inhibit Aβ-induced rods in both rodent and human neurons with effective concentrations for 50% rod reduction (EC50) of 1–10 nM. Remarkably, two D-amino acid receptor-active peptides (RAP-103 and RAP-310) inhibit Aβ-induced rods with an EC50 of ~1 pM in mouse neurons and ~0.1 pM in human neurons. These peptides are analogs of D-Ala-Peptide T-Amide (DAPTA) and share a pentapeptide sequence (TTNYT) antagonistic to several CCR-dependent responses. RAP-103 does not inhibit neuritogenesis or outgrowth even at 1 µM, >106-fold above its EC50. N-terminal methylation, or D-Thr to D-Ser substitution, decreases the rod-inhibiting potency of RAP-103 by 103-fold, suggesting high target specificity. Neither RAP peptide inhibits neuronal rod formation induced by excitotoxic glutamate, but both inhibit rods induced in human neurons by several PrPC/NOX pathway activators (Aβ, HIV-gp120 protein, and IL-6). Significantly, RAP-103 completely protects against Aβ-induced loss of mature and developing synapses and, at 0.1 nM, reverses rods in both rodent and human neurons (T½ ~ 3 h) even in the continuous presence of Aβ. Thus, this orally available, brain-permeable peptide should be highly effective in reducing rod pathology in multifactorial neurological diseases with mixed proteinopathies acting through PrPC/NOX. Full article
Show Figures

Graphical abstract

25 pages, 15626 KiB  
Article
Characterization of a Human Neuronal Culture System for the Study of Cofilin–Actin Rod Pathology
by Lubna H. Tahtamouni, Sydney A. Alderfer, Thomas B. Kuhn, Laurie S. Minamide, Soham Chanda, Michael R. Ruff and James R. Bamburg
Biomedicines 2023, 11(11), 2942; https://doi.org/10.3390/biomedicines11112942 - 31 Oct 2023
Cited by 2 | Viewed by 2414
Abstract
Cofilactin rod pathology, which can initiate synapse loss, has been extensively studied in rodent neurons, hippocampal slices, and in vivo mouse models of human neurodegenerative diseases such as Alzheimer’s disease (AD). In these systems, rod formation induced by disease-associated factors, such as soluble [...] Read more.
Cofilactin rod pathology, which can initiate synapse loss, has been extensively studied in rodent neurons, hippocampal slices, and in vivo mouse models of human neurodegenerative diseases such as Alzheimer’s disease (AD). In these systems, rod formation induced by disease-associated factors, such as soluble oligomers of Amyloid-β (Aβ) in AD, utilizes a pathway requiring cellular prion protein (PrPC), NADPH oxidase (NOX), and cytokine/chemokine receptors (CCR5 and/or CXCR4). However, rod pathways have not been systematically assessed in a human neuronal model. Here, we characterize glutamatergic neurons differentiated from human-induced pluripotent stem cells (iPSCs) for the formation of rods in response to activators of the PrPC-dependent pathway. Optimization of substratum, cell density, and use of glial-conditioned medium yielded a robust system for studying the development of Aβ-induced rods in the absence of glia, suggesting a cell-autonomous pathway. Rod induction in younger neurons requires ectopic expression of PrPC, but this dependency disappears by Day 55. The quantification of proteins within the rod-inducing pathway suggests that increased PrPC and CXCR4 expression may be factors in the doubling of the rod response to Aβ between Days 35 and 55. FDA-approved antagonists to CXCR4 and CCR5 inhibit the rod response. Rods were predominantly observed in dendrites, although severe cytoskeletal disruptions prevented the assignment of over 40% of the rods to either an axon or dendrite. In the absence of glia, a condition in which rods are more readily observed, neurons mature and fire action potentials but do not form functional synapses. However, PSD95-containing dendritic spines associate with axonal regions of pre-synaptic vesicles containing the glutamate transporter, VGLUT1. Thus, our results identified stem cell-derived neurons as a robust model for studying cofilactin rod formation in a human cellular environment and for developing effective therapeutic strategies for the treatment of dementias arising from multiple proteinopathies with different rod initiators. Full article
Show Figures

Graphical abstract

17 pages, 3013 KiB  
Article
Simufilam Reverses Aberrant Receptor Interactions of Filamin A in Alzheimer’s Disease
by Hoau-Yan Wang, Erika Cecon, Julie Dam, Zhe Pei, Ralf Jockers and Lindsay H. Burns
Int. J. Mol. Sci. 2023, 24(18), 13927; https://doi.org/10.3390/ijms241813927 - 11 Sep 2023
Cited by 15 | Viewed by 7824
Abstract
Simufilam is a novel oral drug candidate in Phase 3 clinical trials for Alzheimer’s disease (AD) dementia. This small molecule binds an altered form of filamin A (FLNA) that occurs in AD. This drug action disrupts FLNA’s aberrant linkage to the α7 nicotinic [...] Read more.
Simufilam is a novel oral drug candidate in Phase 3 clinical trials for Alzheimer’s disease (AD) dementia. This small molecule binds an altered form of filamin A (FLNA) that occurs in AD. This drug action disrupts FLNA’s aberrant linkage to the α7 nicotinic acetylcholine receptor (α7nAChR), thereby blocking soluble amyloid beta1–42 (Aβ42)’s signaling via α7nAChR that hyperphosphorylates tau. Here, we aimed to clarify simufilam’s mechanism. We now show that simufilam reduced Aβ42 binding to α7nAChR with a 10-picomolar IC50 using time-resolved fluorescence resonance energy transfer (TR-FRET), a robust technology to detect highly sensitive molecular interactions. We also show that FLNA links to multiple inflammatory receptors in addition to Toll-like receptor 4 (TLR4) in postmortem human AD brains and in AD transgenic mice: TLR2, C-X-C chemokine receptor type 4 (CXCR4), C-C chemokine receptor type 5 (CCR5), and T-cell co-receptor cluster of differentiation 4 (CD4). These aberrant FLNA linkages, which can be induced in a healthy control brain by Aβ42 incubation, were disrupted by simufilam. Simufilam reduced inflammatory cytokine release from Aβ42-stimulated human astrocytes. In the AD transgenic mice, CCR5–G protein coupling was elevated, indicating persistent activation. Oral simufilam reduced both the FLNA–CCR5 linkage and the CCR5–G protein coupling in these mice, while restoring CCR5′s responsivity to C-C chemokine ligand 3 (CCL3). By disrupting aberrant FLNA–receptor interactions critical to AD pathogenic pathways, simufilam may promote brain health. Full article
(This article belongs to the Special Issue Neurodegenerative Disease: From Molecular Basis to Therapy)
Show Figures

Figure 1

Back to TopTop