1
|
Tsou TC, Yeh SC, Tsai FY, Chen PY. Palmitic acid and lipopolysaccharide induce macrophage TNFα secretion, suppressing browning regulators and mitochondrial respiration in adipocytes. Toxicol Appl Pharmacol 2025; 500:117389. [PMID: 40348028 DOI: 10.1016/j.taap.2025.117389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 05/04/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Obesity and its associated pro-inflammatory activity contribute significantly to metabolic dysfunction. In contrast, browning of white adipose tissue (WAT) generally improves metabolic health. Our prior research suggested that macrophage-derived pro-inflammatory cytokines suppress key regulators of browning-adrenergic receptor β3 (Adrb3) and peroxisome proliferator-activated receptor γ (Pparg)-as well as energy metabolism mediators-insulin receptor substrate 1 (Irs1) and hormone-sensitive lipase (Lipe)-in diet-induced obese mice. To explore this mechanism, we developed an in vitro model using RAW264.7 macrophages and 3T3-L1 adipocytes exposed to palmitic acid (PA) and/or lipopolysaccharide (LPS). PA (200 μM) and LPS (1.0 μg/ml) synergistically promoted M1 polarization of macrophages and secretion of pro-inflammatory cytokines, with tumor necrosis factor-α (TNFα), C-C motif chemokine ligand 2 (CCL2), CCL5, and interleukin-6 (IL-6) being predominant. Conditioned media from both control and PA-treated macrophages, when exposed to LPS ≥0.01 μg/ml, significantly downregulated Adrb3, Pparg, Irs1, and Lipe in adipocytes. At physiologically relevant LPS levels (≤0.001 μg/ml), PA-treated macrophage media exerted greater suppression of these genes than controls. Among the cytokines, TNFα emerged as the primary mediator, significantly reducing expression of the four key regulators. Furthermore, adipocytes treated with TNFα exhibited significant reductions in both uncoupling protein 1 (Ucp1) expression and mitochondrial respiration. These findings demonstrate that exposure to obesity-associated factors (PA and LPS) induces macrophage-derived TNFα, which suppresses browning and mitochondrial function in adipocytes. This mechanism may inform new therapeutic strategies targeting TNFα to alleviate obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Tsui-Chun Tsou
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan.
| | - Szu-Ching Yeh
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - Feng-Yuan Tsai
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - Pei-Yu Chen
- National Institute of Environmental Health Sciences, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| |
Collapse
|
2
|
da Silva EA, Faber J, Penitente AR, Fernandes J, Bertolucci PHF, Longo BM, Arida RM. Effects of resistance exercise on behavioral and molecular changes in transgenic female mice for Alzheimer's disease in early and advanced stages. Exp Neurol 2025; 388:115217. [PMID: 40089002 DOI: 10.1016/j.expneurol.2025.115217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that affects memory and cognition, with a higher prevalence in women. Given the lack of effective treatment, physical activity stands out as a complementary approach to prevent or delay disease progression. While numerous studies on humans and animals indicate that aerobic exercise induces brain changes, the impact of resistance exercise (RE) on AD is not fully understood. OBJECTIVE This study aimed to investigate the behavioral and molecular changes induced by RE in female transgenic mice with AD at the early and advanced stages of the disease. MATERIALS AND METHODS Adult (initial phase - 7 to 8 months of age, n = 32) and adult/elderly (advanced phase - 22 to 23 months of age, n = 32) female mice (2xTg-AD) for the APPSWE/PS1dE9 mutation were subjected to a four-week RE protocol. Mobility, anxiety-like behavior, long-term memory (LTM), and depressive-like behavior were assessed. Beta-amyloid (βA) and cytokines were quantified using the ELISA technique. RESULTS There was a progressive increase in strength in both trained groups at different ages. RE reversed memory deficits only in adult AD animals and the anxiety-like behavior only in adult/elderly AD animals. RE reversed depressive-like behavior in adult and adult/elderly AD animals. RE reduced βA only in adult AD animals. RE modified the expression of several cytokines in animals in the early and advanced stage of AD. CONCLUSION RE can be a promising strategy to minimize the deleterious effects of AD; however, its effectiveness may be more limited to the early stages of the disease.
Collapse
Affiliation(s)
| | - Jean Faber
- Federal University of Sao Paulo - Neurology and Neurosurgery Department, Brazil
| | | | | | | | | | | |
Collapse
|
3
|
Zhang W, Meng L, Zhang X, Li Z, Hong F. CNPY2 drives DSS-induced colitis via the macrophage-ROS axis. Biomed Pharmacother 2025; 187:118078. [PMID: 40262233 DOI: 10.1016/j.biopha.2025.118078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal tract, driven by genetic, environmental, and immune system factors. However, its exact mechanisms remain unclear. Here, we demonstrate that CNPY2 plays a critical role in colitis by modulating macrophage activity. Mice with whole-body Cnpy2 knockout (KO) exhibited significantly reduced dextran sodium sulfate (DSS)-induced colitis compared to wild-type controls. Cnpy2 KO mice showed less mucosal barrier disruption and fewer lamina propria macrophages (LPMφs) following DSS treatment. Proinflammatory cytokine production was also diminished in the colons of Cnpy2 KO mice. Furthermore, Cnpy2 KO macrophages generated markedly lower levels of reactive oxygen species (ROS), partly through CHOP regulation. Notably, treatment with the ROS scavenger N-acetyl-L-cysteine (NAC) completely abolished DSS-induced colitis in Cnpy2 KO mice. Thus, CNPY2 exacerbates DSS-induced colitis primarily through macrophage-specific effects, with ROS upregulation being central to its pathogenic role.
Collapse
Affiliation(s)
- Wenqing Zhang
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, United States; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH 43210, United States
| | - Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH 43210, United States
| | - Xiaoli Zhang
- Biostatistics Core, College of Nursing, College of Public Health, University of South Florida, 4202 E Fowler Ave, Tampa, FL 33620, United States
| | - Zihai Li
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, United States; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH 43210, United States.
| | - Feng Hong
- Pelotonia Institute for Immune-Oncology, The Ohio State University Comprehensive Cancer Center, United States; Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, 410 W 12th Ave, Columbus, OH 43210, United States.
| |
Collapse
|
4
|
Toba H, Jin D, Takai S. Suppressing SPARC gene with siRNA exerts therapeutic effects and inhibits MMP-2/9 and ADAMTS1 overexpression in a murine model of ischemia/reperfusion-induced acute kidney injury. J Pharmacol Sci 2025; 158:103-112. [PMID: 40288820 DOI: 10.1016/j.jphs.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC), a collagen-binding matricellular protein, is reported to facilitate inflammation and fibrosis in various tissues including the kidneys. Ischemia/reperfusion (I/R) is a major process of acute kidney injury. To investigate whether SPARC inhibition might attenuate renal I/R injury, we injected small interfering RNA (siRNA) targeting SPARC into male BALB/c mice one day before 45 min of renal ischemia followed by 72 h of reperfusion. Serum creatinine concentration, blood urea nitrogen, histological tubular damage, tubulointerstitial fibrosis, and expression of collagen I and transforming growth factor-β were increased after I/R. Expression of 4-hydroxy-2-nonenal, an oxidative stress marker, and the inflammatory cytokines monocyte chemoattractant protein-1 and tumor necrosis factor-α, were also upregulated in I/R kidneys. Overexpression of SPARC mRNA was observed after I/R, and immunohistochemistry revealed that SPARC was localized mainly in damaged tubuloepithelial cells. Additionally, a disintegrin and metalloproteinase with thrombospondin type 1 motif (ADAMTS1) expression colocalized with SPARC. Injection of siRNA targeting SPARC attenuated renal dysfunction, histological abnormalities, collagen deposition, oxidative stress, and renal inflammation. In addition, SPARC gene knockdown suppressed the I/R-induced increases in ADAMTS1 and matrix metalloproteinase-2/9 expression. In conclusion, I/R-induced SPARC could be a novel therapeutic target against acute kidney injury.
Collapse
Affiliation(s)
- Hiroe Toba
- Laboratory of Clinical Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, 1 Misasagi Shichono-cho, Yamashina-ku, Kyoto, 607-8412, Japan; Department of Pharmacology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki-City, Osaka, 569-8686, Japan.
| | - Denan Jin
- Department of Pharmacology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki-City, Osaka, 569-8686, Japan
| | - Shinji Takai
- Department of Pharmacology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki-City, Osaka, 569-8686, Japan
| |
Collapse
|
5
|
Lai WY, Chuang CW, Huang YC, Huang CJ. Therapeutic Potential of Plant-Derived Small Extracellular Vesicles in Sepsis: A Network Meta-analysis. Pharmacol Res 2025:107795. [PMID: 40414583 DOI: 10.1016/j.phrs.2025.107795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/22/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Sepsis is a life-threatening condition characterized by systemic inflammation and multi-organ dysfunction. Plant-derived small extracellular vesicles (sEVs) have emerged as promising therapeutic agents due to their antioxidant, anti-inflammatory, and immunomodulatory properties. This study conducted a network meta-analysis to identify the most effective plant-derived sEVs for reducing sepsis-induced inflammation and oxidative stress. The analysis included 13 studies evaluating 10 plant-derived sEVs in sepsis-mimicking conditions, with primary outcomes focused on cytokine levels and reactive oxygen species (ROS) production in vitro and in vivo. Secondary outcomes included nuclear factor erythroid 2-related factor 2 (Nrf2) expression and cell viability. The study protocol was registered with PROSPERO (CRD420251011005). Ginger-derived sEVs were identified as the most effective, significantly reducing pro-inflammatory cytokines (interleukin-6 and tumor necrosis factor-α), increasing the anti-inflammatory cytokine (interleukin-10), and suppressing ROS production. They also enhanced Nrf2 expression and improved cell viability, highlighting their role in antioxidant defense and cytoprotection. In conclusion, ginger-derived sEVs are the most effective plant-derived sEVs for mitigating sepsis-induced inflammation and oxidation in both in vitro and in vivo sepsis-mimicking models.
Collapse
Affiliation(s)
- Wen-Yi Lai
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ching-Wei Chuang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chen Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Dermatology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Jen Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
6
|
Thanh Loc V, Bolor T, Purevdorj E, Solongo A, Xuan Ha N, Huong TT, Khanh PN, Thao DT, Pham-The H, Cuong NM. Therapeutic effects of Chrysanthemum zawadzkii and its active compound eupatilin in rheumatoid arthritis: in silico and in vitro validation. Nat Prod Res 2025:1-8. [PMID: 40401641 DOI: 10.1080/14786419.2025.2508835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 05/12/2025] [Accepted: 05/14/2025] [Indexed: 05/23/2025]
Abstract
The 50% ethanol extract of the aerial parts of Chrysanthemum zawadzkii (CZ extract) was evaluated for its inhibitory effects on the production of NO, TNF-α, and XO, and seven isolated flavonoids were tested for COX-2 inhibitory activity. The ethanol extract of C. zawadzkii exhibited a strong inhibitory effect on NO and XO overproduction, with IC50 values of 6.91 µg/mL and 83.48 µg/mL, respectively. Eupatilin (3) significantly suppressed COX-2 expression at a concentration of 100 µM (p < 0.05). Molecular docking analysis and ADMET predictions revealed that eupatilin forms hydrogen bonds and π-σ interactions, which significantly contribute to its binding affinity with residues in the enzyme's active site. Additionally, it exhibits low oral toxicity and good bioavailability in humans. These findings demonstrate the potential of CZ extract and its active compound, eupatilin, as naturally occurring agents with anti-heumatoid arthritis activities, capable of potently inhibiting COX-2 signalling in vitro.
Collapse
Affiliation(s)
| | - Tsolmon Bolor
- Institute of Chemistry and Chemical Technology (MAS), Ulaanbaatar, Mongolia
| | | | - Amgalan Solongo
- Institute of Chemistry and Chemical Technology (MAS), Ulaanbaatar, Mongolia
| | | | | | | | | | - Hai Pham-The
- Department of Life Sciences, University of Science and Technology of Hanoi (VAST), Hanoi, Vietnam
| | - Nguyen Manh Cuong
- Institute of Chemistry (VAST), Hanoi, Vietnam
- Faculty of Chemistry, Graduate University of Science and Technology (VAST), Hanoi, Vietnam
| |
Collapse
|
7
|
El-Gedamy MS, Elnagar SS, Shabana ESE, Derbala SA. Association of Tumor Necrosis Factor Alpha Gene Polymorphism (TNF-α 308 G/A) with Enhanced Severity of Metal-Induced Chronic Kidney Disease via Upregulation of Transmembrane Protein Expression. Biol Trace Elem Res 2025:10.1007/s12011-025-04650-w. [PMID: 40358912 DOI: 10.1007/s12011-025-04650-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025]
Abstract
Chronic kidney disease (CKD) is a progressive disorder often aggravated by environmental exposure to waterborne metal pollutants, such as lead (Pb), cobalt (Co), copper (Cu), and zinc (Zn).These metals induce renal injury predominantly through oxidative stress and inflammatory pathways, with tumor necrosis factor-alpha (TNF-α) serving as a key mediator. The study aimed to investigate the association of the TNF-α -308 G/A (rs1800629) gene polymorphism and transmembrane(Tm)-TNF-α expression with the risk and severity of metal-induced CKD. A case-control study was conducted, including 80 patients with CKD (40 with early-stage and 40 with late-stage disease) and 40 age- and sex-matched healthy controls, selected according to stringent inclusion criteria. Metal concentrations were quantified using inductively coupled plasma mass spectrometry (ICP-MS), TNF-α genotyping was performed via amplification refractory mutation system-polymerase chain reaction (ARMS-PCR), and Tm-TNF-α levels were assessed by flow cytometry. The findings revealed that the A allele was significantly associated with increased CKD risk across dominant, recessive, and allelic genetic models. In a co-dominant model, the AA genotype conferred a fourfold and 15-fold higher risk for early- and late-stage CKD, respectively, compared to those with the GG genotype. Moreover, elevated Tm-TNF-α expression was significantly associated with the GA and AA genotypes and was most pronounced in late-stage CKD patients (p < 0.001). These findings suggest that the TNF-α -308 G/A polymorphism markedly enhances susceptibility to metal-induced CKD, driven by increased Tm-TNF-α expression. Understanding this genetic variation elucidates the pathogenesis of metal-induced nephropathy and offers new opportunities for early diagnosis and targeted therapeutic development.
Collapse
Affiliation(s)
- Mohammed S El-Gedamy
- Department of Medical Laboratories Technology, College of Health and Medical Technologies, Al-Ayen Iraqi University (AUIQ), Thi-Qar, 64001, Iraq.
- Department of Clinical Biochemistry and Molecular Biology, Urology and Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Sherouk S Elnagar
- Department of Internal Medicine, Nephrology Unit, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - El-Shaimaa E Shabana
- Unit of Genetics, Faculty of Medicine, University Pediatrics Hospital, Mansoura University, Mansoura, 35516, Egypt
| | - Safaa A Derbala
- Department of Clinical Biochemistry and Molecular Biology, Urology and Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
8
|
Kireev FD, Lopatnikova JA, Alshevskaya AA, Sennikov SV. Role of Tumor Necrosis Factor in Tuberculosis. Biomolecules 2025; 15:709. [PMID: 40427602 PMCID: PMC12108764 DOI: 10.3390/biom15050709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/28/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Tumor necrosis factor (TNF) is a key immunoregulatory cytokine with a dual role in the host response to Mycobacterium tuberculosis. While essential for granuloma formation, macrophage activation, and containment of latent infection, TNF can also contribute to tissue damage and immune pathology. This review systematically analyzes over 300 peer-reviewed studies published between 1980 and 2024, highlighting the molecular and cellular mechanisms of TNF action in tuberculosis (TB). Particular attention is given to TNF receptor signaling pathways, the balance between protective and pathological immune responses, and the modulation of TNF activity during anti-TNF therapy in patients with autoimmune diseases. We discuss how different TNF inhibitors vary in their capacity to interfere with host defense mechanisms, with monoclonal antibodies carrying a higher reactivation risk than receptor-based agents. To enhance conceptual clarity, we provide newly developed schematic representations that integrate current knowledge on TNF-driven immune dynamics, including its interaction with other cytokines, effects on granuloma stability, and role in intracellular bacterial control. Understanding the pleiotropic functions of TNF in tuberculosis pathogenesis is crucial for developing safe immunomodulatory strategies and optimizing the clinical management of patients at risk of latent TB reactivation.
Collapse
Affiliation(s)
- Fedor D. Kireev
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution “Research Institute of Fundamental and Clinical Immunology” (RIFCI), 630099 Novosibirsk, Russia; (F.D.K.); (J.A.L.)
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education “I.M. Sechenov First Moscow State Medical University” under the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia;
| | - Julia A. Lopatnikova
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution “Research Institute of Fundamental and Clinical Immunology” (RIFCI), 630099 Novosibirsk, Russia; (F.D.K.); (J.A.L.)
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education “I.M. Sechenov First Moscow State Medical University” under the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia;
| | - Alina A. Alshevskaya
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education “I.M. Sechenov First Moscow State Medical University” under the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia;
| | - Sergey V. Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution “Research Institute of Fundamental and Clinical Immunology” (RIFCI), 630099 Novosibirsk, Russia; (F.D.K.); (J.A.L.)
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education “I.M. Sechenov First Moscow State Medical University” under the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia;
| |
Collapse
|
9
|
Fusco A, Guarnieri A, Scieuzo C, Triunfo M, Salvia R, Donnarumma G, Falabella P. Hermetia illucens-Derived Chitosan: A Promising Immunomodulatory Agent for Applications in Biomedical Fields. Biomacromolecules 2025; 26:3224-3233. [PMID: 40300853 PMCID: PMC12076490 DOI: 10.1021/acs.biomac.5c00362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/01/2025]
Abstract
Chitosan, renowned for its important biological properties, is a valuable pharmaceutical excipient for different therapeutic approaches. Currently, the demand for the biopolymer on the market is growing, and, for this reason, it is important to biologically characterize the biopolymer produced from an alternative source to crustaceans, specifically the bioconverter insect Hermetia illucens. In this work, insect chitosan, yielded via heterogeneous and homogeneous deacetylation from larvae, pupal exuviae, and adults, was studied as an immunomodulatory agent. The inflammatory response of immortalized human keratinocyte cells was induced by Salmonella enterica subsp. enterica serovar Typhimurium lipopolysaccharide. After that, the ability of the biopolymer to reduce the expression of the pro-inflammatory cytokines IL-6, IL-8, IL-1α, and TNF-α was tested after 6 and 24 h of treatment. Insect chitosan samples effectively downregulated cytokine expression, with improved activity obtained from heterogeneous chitosan treatments.
Collapse
Affiliation(s)
- Alessandra Fusco
- Department
of Life Sciences, Health and Health Professions, Link Campus University, 00165 Rome, Italy
- Department
of Experimental Medicine, University of
Campania “Luigi Vanvitelli”, Naples 80138, Italy
| | - Anna Guarnieri
- Department
of Basic and Applied Sciences, University
of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Carmen Scieuzo
- Department
of Basic and Applied Sciences, University
of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
- Spinoff
XFlies s.r.l, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Micaela Triunfo
- Department
of Basic and Applied Sciences, University
of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Rosanna Salvia
- Department
of Basic and Applied Sciences, University
of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
- Spinoff
XFlies s.r.l, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Giovanna Donnarumma
- Department
of Experimental Medicine, University of
Campania “Luigi Vanvitelli”, Naples 80138, Italy
| | - Patrizia Falabella
- Department
of Basic and Applied Sciences, University
of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
- Spinoff
XFlies s.r.l, University of Basilicata, Via dell’Ateneo Lucano 10, 85100 Potenza, Italy
| |
Collapse
|
10
|
Srivastava V, O'Reilly C. Characteristics of cerebrospinal fluid in autism spectrum disorder - A systematic review. Neurosci Biobehav Rev 2025; 174:106202. [PMID: 40354953 DOI: 10.1016/j.neubiorev.2025.106202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 04/05/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Autism Spectrum Disorder (ASD) is a range of neurodevelopmental conditions characterized by impaired social interaction, learning, and restricted or repetitive behaviors. The underlying causes of ASD are still debated, but researchers have found many physiological traits like gut problems and impaired immune system to help understand the etiology of ASD. Cerebrospinal fluid (CSF) plays a critical role in maintaining the homeostasis of the neuronal environment and has, therefore, been analyzed in multiple conditions impacting the central nervous system. The study of CSF is crucial to understanding neurological disorders as its composition changes with the disorders, and these changes may indicate various disorder-related physiological mechanisms. For this systematic review, we searched PubMed, Scopus, and Web of Science for studies published between 1977 and 2025 and selected 49 studies after manual screening. We took stock of the evidence supporting the hypothesis that ASD alters the properties and composition of CSF. We systematically report on the different attributes of CSF in the ASD population that could be potential biomarkers and assist in understanding the origins and progression of ASD. We found that in CSF, immune markers, proteins, extra-axial CSF, folate, oxytocin, and vasopressin showed changes in ASD compared to the neurotypicals. We observed gaps in the literature due to variations in age and sample size and noted biases related to sex (i.e., samples are predominantly including male participants) and age (i.e., a handful of studies were conducted on adults). Our review highlights the need for more research on CSF in ASD to improve our understanding of this disorder and identify CSF biomarkers.
Collapse
Affiliation(s)
- Vandana Srivastava
- AI Institute, University of South Carolina, 5th floor, 1112 Greene St., Columbia, SC 29201, USA; Department of Computer Science and Engineering, University of South Carolina, 550 Assembly Street, Columbia, SC 29201, USA; Carolina Autism and Neurodevelopment Research Center, University of South Carolina, 1800 Gervais Street, Columbia, SC 29201, USA.
| | - Christian O'Reilly
- AI Institute, University of South Carolina, 5th floor, 1112 Greene St., Columbia, SC 29201, USA; Department of Computer Science and Engineering, University of South Carolina, 550 Assembly Street, Columbia, SC 29201, USA; Carolina Autism and Neurodevelopment Research Center, University of South Carolina, 1800 Gervais Street, Columbia, SC 29201, USA; Institute for Mind and Brain, University of South Carolina, 1800 Gervais Street, Columbia, SC 29201, USA.
| |
Collapse
|
11
|
Peng G, Li B, Han H, Yuan Y, Mishra F, Huang Y, Liu ZR. Extracellular PKM2 modulates cancer immunity by regulating macrophage polarity. Cancer Immunol Immunother 2025; 74:195. [PMID: 40343475 PMCID: PMC12064527 DOI: 10.1007/s00262-025-04050-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/10/2025] [Indexed: 05/11/2025]
Abstract
Tumor controls its immunity by educating its microenvironment, including regulating polarity of tumor associated macrophages. It is well documented that cancer cells release PKM2 to facilitate tumor progression. We report here that the extracellular PKM2 (EcPKM2) modulates tumor immunity by facilitating M2 macrophage polarization in tumors. EcPKM2 interacts with integrin αvβ3 on macrophage to activate integrin-FAK-PI3K signal axis. Activation of FAK-PI3K by EcPKM2 suppresses PTEN expression, which subsequently upregulates arginase1 (Arg1) expression and activity in macrophage to facilitate M2 polarity. Our studies uncover a novel and important mechanism for modulation of tumor immunity. More importantly, an antibody against PKM2 that disrupts the interaction between EcPKM2 and integrin αvβ3 is effective in converting M2 macrophages to M1 macrophages in tumors, suggesting a new therapeutic strategy and target for cancer therapies. Combination of the anti-PKM2 antibody with checkpoint blockades provides enhanced treatment effects.
Collapse
Affiliation(s)
- Guangda Peng
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA
| | - Bin Li
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA
| | - Hongwei Han
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA
| | - Yi Yuan
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA
| | - Falguni Mishra
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA
| | - Yang Huang
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA
| | - Zhi-Ren Liu
- Department of Biology, Georgia State University, University Plaza, Atlanta, GA, 30303, USA.
| |
Collapse
|
12
|
Dogra A, Savitt AG, Ghebrehiwet B. C1q Binds to CD4+ T Cells and Inhibits the Release of Pro-Inflammatory Cytokines: Role in the Pathogenesis of Systemic Lupus Erythematosus. Int J Mol Sci 2025; 26:4468. [PMID: 40429614 PMCID: PMC12111149 DOI: 10.3390/ijms26104468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
The association between C1q deficiency and the development of Systemic Lupus Erythematosus (SLE) is well established. Several studies have shown that deficiency in C1q is associated with failed apoptotic cleanup, leading to SLE progression. However, the magnitude of this correlation indicates that C1q may play a much more complex role in the development of lupus. This study provides further insight into the pathogenesis of SLE by investigating the consequences of the interaction between C1q and CD4+ T-cells in the breakdown of self-tolerance. Since the C1q/C1q receptor interaction is postulated to play a role, we first confirmed the presence of surface-expressed C1q and C1q receptors on CD4+ T-cells. Then, cell proliferation assays were performed in the presence and absence of purified C1q, gC1qR, and cC1qR. The supernatants of these cultures were used to determine the levels of immunoregulatory cytokines released. Our data confirm that increasing concentrations of C1q and gC1qR significantly inhibited cell proliferation. Furthermore, the CD4+ cells treated with either C1q or gC1qR secreted reduced inflammatory cytokines, such as IL-6 and TNF-alpha, compared to the untreated controls, suggesting that C1q deficiency facilitates the uncontrolled secretion of these critical cytokines, thus contributing to SLE. Although the role of pro-inflammatory cytokines in the induction of SLE is well documented, the mechanism by which C1q contributes to the disease is still a study in progress. Our data demonstrate that the interaction between C1q and its receptors on CD4+ T cells plays a critical role in the suppression of pro-inflammatory cytokines that cause tissue injury in SLE. Therefore, the C1q-C1qR axis may provide a rationally sound target for the design of novel therapeutic approaches for SLE treatment.
Collapse
Affiliation(s)
- Arushi Dogra
- Division of Rheumatology, Allergy and Clinical Immunology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Anne G. Savitt
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Berhane Ghebrehiwet
- Division of Rheumatology, Allergy and Clinical Immunology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
13
|
Thilakan AT, Nandakumar N, Menon RS, Nair SV, Shenoy V, Sathy BN. The impact of size scales and orientations of polymeric scaffold architectural cues on human macrophage polarisation. Biomed Mater 2025; 20:035026. [PMID: 40273929 DOI: 10.1088/1748-605x/add06e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 04/24/2025] [Indexed: 04/26/2025]
Abstract
Macrophage polarisation is crucial for initiating inflammation in response to biomaterial scaffolds, significantly influencing tissue integration and regenerationin vivo. Modulating macrophage polarisation towards a tissue-regeneration-favouring phenotype through the physical properties of scaffolds offers a promising strategy to enhance tissue regeneration while minimising unfavourable immune responses. However, the critical impact of scaffold physical properties, such as size-scale dimensions, orientation of architectural cues, and local-stiffness of these cues on macrophage polarisation, remains largely unexplored and inadequately understood. This study investigates the combinatorial effects of the physical properties of 3D scaffolds made from poly (ϵ-caprolactone) on human macrophage polarisation. Our findings indicate that the size-scale dimensions and orientation of the architectural cues of the scaffold play crucial roles in determining cell shape, attachment, and the modulation of key gene expression (iNOS, IL-1β, MRC1, ARG), as well as cytokines (TNF-α, IL-10) release associated with the polarisation of human macrophages. Specifically, in scaffolds with architectural cues at larger scales (⩾300 µm diameter), macrophage polarisation is primarily determined by the size-scale of the architectural cues and scaffold stiffness, rather than orientation. Conversely, at smaller scales (⩽15 µm), the orientation of the scaffold's architectural cues plays a more critical role. These insights underscore the pivotal role of scaffold design in modulating immune responses for enhanced tissue regeneration, offering valuable guidance for the rational development of biomaterial scaffolds in regenerative medicine.
Collapse
Affiliation(s)
- Akhil T Thilakan
- Amrita School of Nanosciences and Molecular medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Niji Nandakumar
- Amrita School of Nanosciences and Molecular medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Revathy S Menon
- Amrita School of Nanosciences and Molecular medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Shantikumar V Nair
- Amrita School of Nanosciences and Molecular medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Veena Shenoy
- Department of Transfusion Medicine, Amrita Institute of Medical Sciences and Research Centre, Kochi, Kerala, India
| | - Binulal Nelson Sathy
- Amrita School of Nanosciences and Molecular medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| |
Collapse
|
14
|
Alcalde-Estévez E, Moreno-Piedra A, Asenjo-Bueno A, Martos-Elvira M, de la Serna-Soto M, Ruiz-Ortega M, Olmos G, López-Ongil S, Ruiz-Torres MP. Aging-related hyperphosphatemia triggers the release of TNF-α from macrophages, promoting indicators of sarcopenia through the reduction of IL-15 expression in skeletal muscle. Life Sci 2025; 368:123507. [PMID: 40010633 DOI: 10.1016/j.lfs.2025.123507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 02/11/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
AIMS The association between aging-related hyperphosphatemia and sarcopenia has been documented, and evidence suggests that inflammaging is involved in the manifestation of sarcopenia. The present study investigates whether hyperphosphatemia triggers inflammation, thereby inducing the appearance of sarcopenia along with the cytokines involved in these processes. MATERIALS AND METHODS RAW 264.7 macrophages were incubated with β-glycerophosphate (BGP), as a phosphate donor, at different time intervals, to assess the production of proinflammatory markers. Conditioned medium from macrophages was collected and added to cultured C2C12 myoblasts to analyse whether proinflammatory molecules, released by macrophages, modified myogenic differentiation, cell senescence or myokine IL-15 expression. A neutralising antibody anti-TNF-α and recombinant IL-15 were added to evaluate the role of these cytokines in the observed effects. Additionally, TNF-α, IL-15, serum phosphate, and sarcopenia signs were evaluated in 5-month-old mice, 24-month-old mice and 24-month-old mice fed with a hypophosphatemic diet. KEY FINDINGS BGP increased TNF-α expression in macrophages through NFkB activation. Conditioned medium from BGP-treated macrophages impaired myogenic differentiation in differentiating myoblasts and promoted cellular senescence and reduced IL-15 expression in undifferentiated myoblasts. These effects were mediated by TNF-α. Old mice displayed reduced expression of muscle IL-15 and elevated circulating TNF-α, along with increased serum phosphate levels, which correlated with the appearance of sarcopenia indicators. The hypophosphatemic diet prevented these changes in old mice. SIGNIFICANCE Hyperphosphatemia induces TNF-α production in macrophages, which contributes to the reduced expression of muscular IL-15. This mechanism may play a role in inducing sarcopenia in elderly mice.
Collapse
Affiliation(s)
- Elena Alcalde-Estévez
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain
| | - Ariadna Moreno-Piedra
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain; Ramón y Cajal Health Research Institute (IRYCIS), Madrid 28034, Spain
| | - Ana Asenjo-Bueno
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain
| | - María Martos-Elvira
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain
| | - Mariano de la Serna-Soto
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain
| | - Marta Ruiz-Ortega
- Cellular and Molecular Biology in Renal and Vascular Pathology, Institute of Medical Research of the Jiménez Díaz Foundation, Autonomous University of Madrid, Madrid, Spain
| | - Gemma Olmos
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain; Renal and Vascular Physiology and Physiopathology Research Group of Area 5 of IRYCIS, Madrid 28034, Spain; Reina Sofía Institute of Nephrology Research (IRSIN), Íñigo Álvarez de Toledo Renal Foundation (FRIAT), Madrid 28003, Spain
| | - Susana López-Ongil
- Renal and Vascular Physiology and Physiopathology Research Group of Area 5 of IRYCIS, Madrid 28034, Spain; Reina Sofía Institute of Nephrology Research (IRSIN), Íñigo Álvarez de Toledo Renal Foundation (FRIAT), Madrid 28003, Spain; Foundation for Biomedical Research of the Príncipe de Asturias University Hospital, Alcalá de Henares, Madrid 28805, Spain
| | - María P Ruiz-Torres
- University of Alcalá, Faculty of Medicine and Health Sciences, Department of Systems Biology, Alcalá de Henares, Madrid 28871, Spain; Renal and Vascular Physiology and Physiopathology Research Group of Area 5 of IRYCIS, Madrid 28034, Spain; Reina Sofía Institute of Nephrology Research (IRSIN), Íñigo Álvarez de Toledo Renal Foundation (FRIAT), Madrid 28003, Spain
| |
Collapse
|
15
|
Abianeh HS, Kesharwani P, Sahebkar A. The use of aptamers as therapeutic inhibitors and biosensors of TNF-alpha. Int J Biol Macromol 2025; 306:141202. [PMID: 39971069 DOI: 10.1016/j.ijbiomac.2025.141202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/13/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025]
Abstract
Tumor necrosis factor-alpha (TNF-α) is a pivotal cytokine in the pathogenesis of numerous inflammatory and autoimmune diseases. Precise and sensitive detection of TNF-α is essential for both clinical applications and research endeavors. In the realm of cytokine detection, particularly TNF-α, the development of highly sensitive and specific biosensors has become a focal point. The biosensing landscape encompasses a variety of biorecognition elements, each with its unique set of characteristics. TNF inhibitors come with a significant price tag and, notably, do not yield positive responses in all patients. Despite the availability of numerous FDA-approved biologic agents (e.g., infliximab, adalimumab, certolizumab pegol, etc.) and monoclonal antibodies (e.g., adalimumab) targeting TNF-α, aptamers tailored for blocking TNF-α activities have yet to receive approval. Aptamers have rapidly gained recognition as readily available, versatile, and highly effective molecular tools for both therapeutic and diagnostic purposes in the context of TNF-alpha. In this manuscript, we explore the potential of short single-stranded DNA or RNA sequences known as aptamers as biorecognition elements in biosensors designed for the detection of TNF-α. We delve into the progress made in the development of aptamer-based TNF-α inhibitors and shed light on successful studies in this burgeoning field.
Collapse
Affiliation(s)
- Hossein Samiei Abianeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
16
|
Solongo T, Huong TT, Purevdorj E, Solongo A, Bayasgalan B, Loc VT, Ha NX, Ha VT, Hung NP, Thao DT, Nga NT, The HP, Stark P, Cuong NM. Leptomonines A and B, two novel rare benzyltetrahydroisoquinoline N-oxides from the aerial parts of Leptopyrum fumarioides as potential COX-2 inhibitors: in vitro and in silico studies. J Nat Med 2025; 79:517-529. [PMID: 40053248 DOI: 10.1007/s11418-025-01882-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/03/2025] [Indexed: 05/09/2025]
Abstract
Leptomonines A and B, two novel rare benzyltetrahydroisoquinoline N-oxides, were isolated from the aerial parts of Leptopyrum fumarioides (L.) Reichenb. collected in Tuv province, Mongolia. Their chemical structures, absolute configurations, and conformations were established by 2D-NMR and CD spectral analyses. Leptomonine A (1) can suppress TNF-α production and COX-2 expression in LPS-stimulated RAW 267.4 cells. This compound at a concentration of 100 µM significantly reduced the TNF-α and COX-2 levels by 36.43% and 47.10%, respectively, compared with the negative control. Moreover, leptomonine B (2) remarkably lowers COX-2 levels at the highest concentration. The docking simulations were conducted with the COX-2 enzyme and revealed the binding ability of leptomonine A (1) and leptomonine B (2) with binding energies of - 9.03 and - 8.96 kcal/mol, respectively. The interactions of these alkaloids with the targets were mainly with the hydrophobic and hydrophilic sites, which are quite similar to rofecoxib. Phytochemical investigation revealed the diversity and novelty of the natural isoquinoline alkaloids in Leptopyrum fumarioides. Two new benzyltetrahydroisoquinoline N-oxides were identified as the bioactive constituents of Leptopyrum fumarioides by assessing its anti-inflammatory effects. The findings provide scientific justification to support the traditional application of Leptopyrum fumarioides for treating liver diseases associated with inflammation.
Collapse
Affiliation(s)
- Tserendorj Solongo
- Institute of Chemistry and Chemical Technology (MAS), Ulaanbaatar, Mongolia
- School of Pharmacy, Mongolian National University of Medical Sciences (MNUMS), Ulaanbaatar, Mongolia
| | - Tran Thu Huong
- Institute of Natural Products Chemistry (INPC), Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | | | - Amgalan Solongo
- Institute of Chemistry and Chemical Technology (MAS), Ulaanbaatar, Mongolia.
| | - Battsagaan Bayasgalan
- School of Pharmacy, Mongolian National University of Medical Sciences (MNUMS), Ulaanbaatar, Mongolia
| | - Vu Thanh Loc
- Institute of Natural Products Chemistry (INPC), Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Nguyen Xuan Ha
- Institute of Natural Products Chemistry (INPC), Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Vu Thi Ha
- Institute of Natural Products Chemistry (INPC), Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Nguyen Phi Hung
- Institute of Chemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Do Thi Thao
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Nguyen Thi Nga
- Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Hai Pham- The
- University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Pauline Stark
- Program Center MetaCom, Leibniz Institute of Plant Biochemistry (IPB), Halle, Germany
| | - Nguyen Manh Cuong
- Institute of Natural Products Chemistry (INPC), Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam.
| |
Collapse
|
17
|
Rahman MM, Talukder A, Rahi MS, Das PK, Grice ID, Ulett GC, Wei MQ. Evaluation of Immunostimulatory Effects of Bacterial Lysate Proteins on THP-1 Macrophages: Pro-inflammatory Cytokine Response and Proteomic Profiling. J Immunol Res 2025; 2025:2289241. [PMID: 40322557 PMCID: PMC12048194 DOI: 10.1155/jimr/2289241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Bacterial lysate proteins (BLPs) serve as potential immunostimulants, recognized by pattern recognition receptors (PRRs) on immune cells, eliciting a robust immune response. In this study, THP-1 macrophages were treated with varying doses of BLPs derived from Streptococcus pyogenes (SP), Streptococcus agalactiae (SA), and Serratia marcescens (SM). The results showed significant increases (p < 0.05) in pro-inflammatory cytokines such as TNF-α, IL-1β, IL-6, IL-12, granulocyte macrophage-colony stimulating factor (GM-CSF), eotaxin, and macrophage inflammatory protein (MIP)-1α, except for 5 µg of all BLPs for TNF-α and eotaxin, and 5 µg of SP for IL-12 production. No significant differences were found between the corresponding doses of SP and SA or SP and SM, except for GM-CSF in all doses, while SA and SM only showed a difference at the 5 µg dose for GM-CSF. Furthermore, there were no significant differences between the 10 and 20 µg doses of all BLPs, indicating that doses higher than 10 µg do not significantly enhance the pro-inflammatory response. Combination doses of SP + SM and SA + SM did not show significant differences, except for IL-1β, suggesting no synergistic effect. Cytotoxicity was observed to increase with higher BLP concentrations in a dose-dependent manner, with combinations of SP + SM and SA + SM exhibiting greater cytotoxicity than the individual BLPs. Proteomic analysis of BLPs identified immunostimulatory proteins, including heat shock proteins (HSPs; ClpB, DnaK, and GroEL), metabolic enzymes (glyceraldehyde 3-phosphate dehydrogenase (GAPDH), enolase, and arginine deiminase (ADI)), and surface and secreted proteins (ESAT-6-like protein, CRISPR-associated endonuclease Cas9, OmpA, porin OmpC, and serralysin), which are involved in immune modulation, bacterial clearance, and immune evasion. This study underscores the potential of bacterial proteins as vaccine adjuvants or supplementary therapies; however, further research is essential to find a balance between immune activation and inflammation reduction to develop safer and more effective immunostimulants.
Collapse
Affiliation(s)
- Md. Mijanur Rahman
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Chittagong, Bangladesh
| | - Asma Talukder
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Md. Sifat Rahi
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Plabon Kumar Das
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - I. Darren Grice
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Glen C. Ulett
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Ming Q. Wei
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
18
|
Ajike RA, Afolabi OA, Alabi BA, Ajayi AF, Oyekunle OS, Lawal SK, Olojede SO, Nku-Ekpang OA, Hezekiah OS, Hammed OS. Sequential administration of febuxostat and vitamin E protects against testicular ischemia/reperfusion injury via inhibition of sperm DNA damage in Wistar rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04095-x. [PMID: 40261353 DOI: 10.1007/s00210-025-04095-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/22/2025] [Indexed: 04/24/2025]
Abstract
The pathway of testicular ischemia-reperfusion injury (TIRI) has been shown to involve reactive oxygen species (ROS) generation in the ischemic phase and later phase of reperfusion. This study was therefore designed to investigate the effect of blockage of ROS in the ischemic and reperfusion phases of TIRI. Thirty male Wistar rats were grouped into five groups (n = 6 rats each): sham, torsion + detorsion (TD), febuxostat (FEB)-administered (TFD) group, vitamin E (V)-administered (TDV) group, and FEB and vitamin E-administered (TFDV) group. Blood samples (for inflammatory and hormonal assay), testicular (for oxidative stress and histopathology), and epididymal (for sperm DNA damage and indices) tissues were collected after 3 days of detorsion. The TFD and TFDV groups showed a significant reduction in XO and MDA (p < 0.001; η2 > 0.7), as well as a concomitant increase in CAT, thiols, and SOD levels when compared with the TD group (p < 0.01, η2 > 0.5). The TFD group significantly reduced all inflammatory markers (p < 0.05; η2 = 0.75). The observed increase (p < 0.05; η2 = 0.92) in LH level, in response to a low level of testosterone in the TD group, was significantly raised in TFD and TFDV groups. The observed decrease (p < 0.001) in inhibin level in the TD group was raised (p < 0.05; η2 = 0.90) in the TDV group only. A significant increase (p < 0.001) in sperm DNA damage in the TD group was significantly reduced (p < 0.05; η2 = 0.88) in all the treatment groups while the reduced sperm viability (p < 0.01) in the TD group was increased (p < 0.05) in the TFDV group only. There was an improvement in the testicular cytoarchitecture in the TFD and TFDV groups. This study showed that sequential administration of febuxostat in the ischemic phase of TT and vitamin E in the later phase of reperfusion protects the testes against TIRI via inhibition of oxidative stress, inflammation, and sperm DNA damage.
Collapse
Affiliation(s)
- Richard Adedamola Ajike
- Department of Physiology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Oladele Ayobami Afolabi
- Department of Physiology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| | - Babatunde Adebola Alabi
- Department of Pharmacology & Therapeutics, Bowen University, Iwo, Osun State, Nigeria
- Department of Pharmacology & Therapeutics, Faculty of Medicine and Pharmacy, Kampala International University in Tanzania, Dar Es Salaam, United Republic of Tanzania
| | - Ayodeji Folorunsho Ajayi
- Department of Physiology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Olubunmi Simeon Oyekunle
- Department of Physiology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Sodiq Kolawole Lawal
- School of Nursing, Faculty of Health Sciences, University of Botswana, Gaborone, Botswana
| | - Samuel Oluwaseun Olojede
- Department of Human Biology, Faculty of Health Sciences, Walter Sisulu University, Mthatha, South Africa
| | - Okot-Asi Nku-Ekpang
- Department of Physiology, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Cross River State, Nigeria
| | - Oluwaseun Samuel Hezekiah
- Department of Physiology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Opeyemi Sodiq Hammed
- Department of Physiology, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
19
|
Adamecz DI, Veres É, Papp C, Árva H, Rónavári A, Marton A, Vizler C, Gácser A, Kónya Z, Igaz N, Kiricsi M. Gold and Silver Nanoparticles Efficiently Modulate the Crosstalk Between Macrophages and Cancer Cells. Int J Nanomedicine 2025; 20:4777-4802. [PMID: 40255669 PMCID: PMC12009049 DOI: 10.2147/ijn.s508171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/02/2025] [Indexed: 04/22/2025] Open
Abstract
Background Macrophages, polarized into pro-inflammatory M1 or anti-inflammatory M2 states, are essential cellular elements of innate immunity. In the tumor microenvironment, owing to a paracrine manipulative program by cancerous cells, tumor-associated macrophages (TAMs) evolve, which can shift between M1-like and M2-like phenotypes. Since it is fairly unknown how the promising anticancer agents, silver (AgNPs) and gold nanoparticles (AuNPs) affect the bidirectional communication and reprogramming in the tumor stroma, we examined the behavior, the tumor-supporting functions, and the expression of polarization and functional marker genes of TAMs to reveal how these are modulated upon interaction with nanoparticle-exposed cancer cells. Methods We established co-cultures of murine immortalized J774 or primary bone marrow-derived macrophages with 4T1 breast cancer cells treated with AuNPs or AgNPs or with none of the nanoparticles. We assessed the expression of macrophage polarization and functional markers using RT-qPCR and Proteome Profiler Array and evaluated macrophage migration and matrix metalloproteinase activity by specific assays. Results Protein and mRNA levels of most examined factors - except tumor necrosis factor-alpha - such as C-C-motif chemokine ligands 2 and 22, interleukin-23, inducible nitric oxide synthase, cyclooxygenase-2, the macrophage mannose receptor CD206, transforming growth factor-beta, and chitinase-like-3 protein decreased, and the expression of polarization markers revealed a shift towards M1-like phenotype in macrophages co-cultured with AgNP- or AuNP-treated 4T1 cells. Both nanoparticle treatments reduced the levels and activity of cell migration-related factors, such as C-C motif chemokine ligand 3, matrix metalloproteinases, and suppressed macrophage migration. Conclusion Both AuNPs and AgNPs showed a remarkable ability to influence macrophage-cancer cell communication, suppressed indirectly M2-like TAM polarization, and perturbed the migration behavior of TAMs that is critical for tumor invasion, indicating modulated immunological functions and debilitated cancer-promoting capabilities of TAMs in this microenvironment.
Collapse
Affiliation(s)
- Dóra Izabella Adamecz
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Éva Veres
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
- Department of Biotechnology and Microbiology, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Pathogen Fungi Research Group, University of Szeged, Szeged, Hungary
| | - Csaba Papp
- Department of Biotechnology and Microbiology, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Pathogen Fungi Research Group, University of Szeged, Szeged, Hungary
| | - Hédi Árva
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Andrea Rónavári
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, Hungary
| | - Annamária Marton
- Laboratory of Tumor Immunology and Pharmacology, Centre of Excellence of the European Union, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Csaba Vizler
- Laboratory of Tumor Immunology and Pharmacology, Centre of Excellence of the European Union, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Attila Gácser
- Department of Biotechnology and Microbiology, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Pathogen Fungi Research Group, University of Szeged, Szeged, Hungary
| | - Zoltán Kónya
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, Hungary
| | - Nóra Igaz
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Mónika Kiricsi
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| |
Collapse
|
20
|
Tan X, Sheng R, Li W, Tao Y, Liu Z, Yang N, Hashmi SS, Feng F, Liu F, Ge L. Mannose-Glycated Metal-Phenolic Microcapsules Orchestrate Phenotype Switch of Macrophages for Boosting Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415565. [PMID: 40014038 PMCID: PMC12021090 DOI: 10.1002/advs.202415565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/04/2025] [Indexed: 02/28/2025]
Abstract
Microcapsules are advancing in immunotherapy, with both their core and shell being capable of loading immunoregulatory substances. Notably, microcapsules with intrinsic bioactivities can more directly modulate the immune microenvironment, while current research in this area remains scarce. Herein, immunomodulatory metal-phenolic microcapsules (mMPMs) is developed through the one-step assembly of dopamine-modified hyaluronic acid (HADA) and FeIII onto mannose-glycated bovine serum albumin microbubbles (Man-BSA MBs). Specifically, Man-BSA formed during the early stages of the Maillard reaction is sonicated to produce microbubbles as templates for capsule preparation. Subsequently, HADA is rapidly coated on the templates and coordinates with FeIII to form microcapsules after air escapes from MBs. Mass spectrometry analysis identifies abundant lysine glycation sites on Man-BSA, with the highest glycation site percentage reaching 94.88%. Man-BSA within mMPMs effectively promotes macrophage internalization, induces the accumulation of pro-inflammatory mediators, and thereby results in the M1 polarization of macrophages, as further corroborated by proteomic analysis. Consequently, the compelling anti-tumor effects of mMPMs are demonstrated both in vitro and in vivo. Overall, this work presents an immunomodulatory microcapsule that activates pro-inflammatory phenotype macrophages, which is a promising microcarrier to improve immunotherapeutic effects.
Collapse
Affiliation(s)
- Xin Tan
- State Key Laboratory of Digital Medical EngineeringSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Renwang Sheng
- School of MedicineSoutheast UniversityNanjing210009China
| | - Weikun Li
- State Key Laboratory of Digital Medical EngineeringSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Yinghua Tao
- State Key Laboratory of Digital Medical EngineeringSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Zonghao Liu
- State Key Laboratory of Digital Medical EngineeringSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Ning Yang
- State Key Laboratory of Digital Medical EngineeringSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Syeda Safia Hashmi
- State Key Laboratory of Digital Medical EngineeringSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
| | - Feiling Feng
- Department of Biliary Tract Surgery IShanghai Eastern Hepatobiliary Surgery HospitalNavy Medical UniversityShanghai200438China
| | - Fangzhou Liu
- Department of Head & Neck SurgeryJiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical UniversityNanjing210029China
| | - Liqin Ge
- State Key Laboratory of Digital Medical EngineeringSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjing210096China
- Advanced Ocean Institute of Southeast UniversityNantong226000China
| |
Collapse
|
21
|
Hiti L, Markovič T, Lainscak M, Farkaš Lainščak J, Pal E, Mlinarič-Raščan I. The immunopathogenesis of a cytokine storm: The key mechanisms underlying severe COVID-19. Cytokine Growth Factor Rev 2025; 82:1-17. [PMID: 39884914 DOI: 10.1016/j.cytogfr.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025]
Abstract
A cytokine storm is marked by excessive pro-inflammatory cytokine release, and has emerged as a key factor in severe COVID-19 cases - making it a critical therapeutic target. However, its pathophysiology was poorly understood, which hindered effective treatment. SARS-CoV-2 initially disrupts angiotensin signalling, promoting inflammation through ACE-2 downregulation. Some patients' immune systems then fail to shift from innate to adaptive immunity, suppressing interferon responses and leading to excessive pyroptosis and neutrophil activation. This amplifies tissue damage and inflammation, creating a pro-inflammatory loop. The result is the disruption of Th1/Th2 and Th17/Treg balances, lymphocyte exhaustion, and extensive blood clotting. Cytokine storm treatments include glucocorticoids to suppress the immune system, monoclonal antibodies to neutralize specific cytokines, and JAK inhibitors to block cytokine receptor signalling. However, the most effective treatment options for mitigating SARS-CoV-2 infection remain vaccines as a preventive measure and antiviral drugs for the early stages of infection. This article synthesizes insights into immune dysregulation in COVID-19, offering a framework to better understand cytokine storms and to improve monitoring, biomarker discovery, and treatment strategies for COVID-19 and other conditions involving cytokine storms.
Collapse
Affiliation(s)
- Luka Hiti
- Faculty of Pharmacy, University of Ljubljana, Slovenia
| | | | - Mitja Lainscak
- General Hospital Murska Sobota, Slovenia; Faculty of Medicine, University of Ljubljana, Slovenia
| | | | - Emil Pal
- General Hospital Murska Sobota, Slovenia
| | | |
Collapse
|
22
|
Ngalula S, Carlin CR. TNF-α-Driven Changes in Polarized EGF Receptor Trafficking Facilitate Phosphatidylinositol 3-Kinase/Protein Kinase B Signaling From the Apical Surface of MDCK Epithelial Cells. Traffic 2025; 26:e70005. [PMID: 40324787 PMCID: PMC12052438 DOI: 10.1111/tra.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/15/2025] [Accepted: 03/14/2025] [Indexed: 05/07/2025]
Abstract
This manuscript describes a novel unconventional secretory pathway that facilitates EGF receptor (EGFR) signaling from apical membranes in polarized epithelial cells responding to immune cell mediators. Epithelial tissues provide a physical barrier between our bodies and the external environment and share an intimate relationship with circulating and local immune cells. Our studies describe an unexpected connection between the proinflammatory cytokine tumor necrosis factor-alpha (TNF-α) and EGFR typically localized to basolateral membranes in polarized epithelial cells. These two molecules sit atop complex biological networks with a long history of shared investigative interest from the vantage point of signaling pathway interactions. We have discovered that TNF-α alters the functional landscape of fully polarized epithelial cells by changing the speed and direction of EGFR secretion. Our results show apical EGFR delivery occurs within minutes of de novo synthesis likely via a direct route from the endoplasmic reticulum without passage through the Golgi complex. Additionally, our studies have revealed that apical cellular compartmentalization constitutes an important mechanism to specify EGFR signaling via phosphatidylinositol-4,5-bisphosphate 3-kinase/protein-kinase-B pathways. Our study paves the way for a better understanding of how inflammatory cytokines fine-tune local homeostatic and inflammatory responses by altering the spatial organization of epithelial cell signaling systems.
Collapse
Affiliation(s)
- Syntyche Ngalula
- Department of Molecular Biology and Microbiology, School of MedicineCase Western Reserve UniversityClevelandOhioUSA
| | - Cathleen R. Carlin
- Department of Molecular Biology and Microbiology, School of MedicineCase Western Reserve UniversityClevelandOhioUSA
- Case Western Reserve University Comprehensive Cancer Center, School of MedicineCase Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
23
|
Leinweber B, Pilorz V, Olejniczak I, Skrum L, Begemann K, Heyde I, Stenger S, Sadik CD, Oster H. Bmal1 deficiency in neutrophils alleviates symptoms induced by high-fat diet. iScience 2025; 28:112038. [PMID: 40124497 PMCID: PMC11930374 DOI: 10.1016/j.isci.2025.112038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/24/2024] [Accepted: 02/12/2025] [Indexed: 03/25/2025] Open
Abstract
Physiological processes, including metabolism and immune responses, are generated by the circadian clock, driven by clock genes. Disrupting circadian rhythms through a high-fat diet promotes obesity and inflammation. Studies show that deleting the clock gene, brain, and muscle ARNT-like 1 (Bmal1) in adipose tissue leads to overeating and weight gain. We now show that Bmal1 deletion in neutrophils protects against diet-induced obesity and reduces inflammatory macrophage infiltration into epididymal white adipose tissue (eWAT), despite increased food intake over 20 weeks of a high-fat diet. This protection is linked to enhanced energy expenditure, increased UCP1 expression in iBAT, improved insulin sensitivity, and altered expression of genes encoding chemokine receptors CXCR2, CXCR4, and the ligand Cxcl2 in eWAT. Our findings reveal a key role of Bmal1 in neutrophils in regulating high-fat diet-induced adipose inflammation and emphasize circadian regulation's importance in immuno-metabolic function.
Collapse
Affiliation(s)
- Brinja Leinweber
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Violetta Pilorz
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Iwona Olejniczak
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Ludmila Skrum
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Kimberly Begemann
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Isabel Heyde
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Sarah Stenger
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| | - Christian David Sadik
- University of Lübeck, Department of Dermatology, Allergy, and Venereology Ratzeburger Allee, 23562 Luebeck, Germany
| | - Henrik Oster
- University of Lübeck, Institute of Neurobiology, Center of Brain, Behaviour and Metabolism, Marie-Curie-Strasse, 23562 Luebeck, Germany
| |
Collapse
|
24
|
Perez-Lopez A, Hernandez-Galicia G, Lopez-Bailon LU, Gonzalez-Telona AD, Rosales-Reyes R, Alpuche-Aranda CM, Santos-Preciado JI, Ortiz-Navarrete V. Pro-inflammatory and anti-inflammatory responses in B cells during Salmonella infection. Eur J Microbiol Immunol (Bp) 2025; 15:32-41. [PMID: 40067375 PMCID: PMC11925187 DOI: 10.1556/1886.2024.00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/12/2025] [Indexed: 03/20/2025] Open
Abstract
B-cells serve as a niche for Salmonella to establish a chronic infection, enabling bacteria to evade immune responses. One mechanism Salmonella uses to survive inside B-cells involves inhibiting the NLRC4 inflammasome activation, thereby preventing pyroptotic cell death. This study investigates whether Salmonella-infected B-cells can mount bactericidal responses to control intracellular bacteria. Our results show that Salmonella-infected B-cells can produce and release TNFα, IL-6, and IL-10, but not RANTES. Furthermore, priming B-cells with TNFα, IL-1β, or IFNγ enhances their bactericidal activity by promoting the production of reactive oxygen and nitrogen production species, reducing intracellular load. These results suggest that B-cells can clear Salmonella infection within a pro-inflammatory environment. However, the concurrent production of IL-10 may counteract the effects of pro-inflammatory cytokines, potentially modulating the immune response in the microenvironment.
Collapse
Affiliation(s)
- Araceli Perez-Lopez
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México. Tlalnepantla, State of Mexico, Mexico
| | - Gabriela Hernandez-Galicia
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Luis Uriel Lopez-Bailon
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Ana D. Gonzalez-Telona
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Roberto Rosales-Reyes
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Celia M. Alpuche-Aranda
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, Mexico
| | - Jose I. Santos-Preciado
- Unidad de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Vianney Ortiz-Navarrete
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
25
|
Kgoadi K, Bajpai P, Ibegbu CC, Dkhar HK, Enriquez AB, Dawa S, Cribbs SK, Rengarajan J. Alveolar macrophages from persons with HIV mount impaired TNF signaling networks to M. tuberculosis infection. Nat Commun 2025; 16:2397. [PMID: 40064940 PMCID: PMC11894076 DOI: 10.1038/s41467-025-57668-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
People living with HIV (PLWH) have an increased risk for developing tuberculosis after M. tuberculosis infection, despite anti-retroviral therapy (ART). To delineate the underlying mechanisms, we conducted single cell transcriptomics on bronchoalveolar lavage cells from PLWH on ART and HIV uninfected healthy controls infected with M. tuberculosis ex vivo. We identify an M1-like proinflammatory alveolar macrophage subset that sequentially acquires TNF signaling capacity in controls but not in PLWH. Cell-cell communication analyses reveal interactions between M1-like macrophages and effector memory T cells within TNF superfamily, chemokine, and costimulatory networks in the airways of controls. These interaction networks were lacking in PLWH infected with M. tuberculosis, where anti-inflammatory M2-like alveolar macrophages and T regulatory cells dominated along with dysregulated T cell signatures. Our data support a model in which impaired TNF-TNFR signaling, M2-like alveolar macrophages and aberrant macrophage-T cell crosstalk, lead to ineffective immunity to M. tuberculosis in PLWH on ART.
Collapse
Affiliation(s)
- Khanyisile Kgoadi
- Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA
| | - Prashant Bajpai
- Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA
| | - Chris C Ibegbu
- Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA
| | | | - Ana Beatriz Enriquez
- Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA
| | - Stanzin Dawa
- Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA
| | - Sushma K Cribbs
- Division of Pulmonary, Allergy, Critical Care & Sleep, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Veterans Affairs, Atlanta, GA, USA.
| | - Jyothi Rengarajan
- Emory Vaccine Center, Emory National Primate Center, Emory University, Atlanta, GA, USA.
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
26
|
Mokgalaboni K. Exploring the anti-inflammatory potential of vitamin D in cardiometabolic diseases. Metabol Open 2025; 25:100348. [PMID: 39876902 PMCID: PMC11773081 DOI: 10.1016/j.metop.2025.100348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
The prevalence of cardiometabolic diseases is rising, and this is fuelled by inflammation, which tends to be worse in individuals with vitamin D (VD) deficiency. While non-steroidal anti-inflammatory interventions are available, they present with coagulation events. Hence, alternative therapy in the form of VD supplements is gaining research interest. This study reviewed the effect of VD supplementation on inflammation, focusing on nuclear factor kappa-beta (NF-κβ), tumour necrosis factor-alpha (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) across different cardiometabolic disease. Thirty-seven studies, 16 rodent models and 21 clinical studies were evaluated. The study considered evidence from rodent models to understand the effect of VD on these markers of inflammation and its translatability to clinical studies. While the potential benefits of VD were notable in rodents, these effects were less consistent in clinical studies. Notably, rodent models showed a more pronounced impact of VD in reducing NF-κβ and TNF-α; however, clinical trials reported conflicting findings. Furthermore, the VD was important in reducing MCP-1 across different rodent models; this was partially demonstrated in clinical trials. Based on these findings, VD modulates inflammation in cardiometabolic disease by inhibiting the activation of NF-κβ and suppressing the production of TNF-α and MCP-1. Although VD has some possible benefits in rodent models, the translatability of these findings in clinical trials is limited. Hence, the presented evidence in this study calls for further randomised controlled trials to assess the effect of VD on inflammation in patients living with different conditions as a therapy to curb the inflammation and the risk thereof. Future trials should also focus on exploring the VD dose-response, optimal dose, and duration of VD intervention among these patients that may offer optimal benefits on inflammation.
Collapse
Affiliation(s)
- Kabelo Mokgalaboni
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Calabash Building, Office no: 02-047 Florida Campus, 1710, South Africa
| |
Collapse
|
27
|
Wu J, Zhang B, Du W, Shi Y, Xie C, Ke Y, Yi X. OC-STAMP is a potential biomarker and therapeutic target for Silicosis: an exploratory investigation. J Transl Med 2025; 23:214. [PMID: 39985047 PMCID: PMC11846239 DOI: 10.1186/s12967-024-05981-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/13/2024] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Silicosis, a disease characterized by fibrous changes in lung tissue due to prolonged silica dust inhalation, exhibits a complex pathogenesis that remains inadequately addressed by current interventions. Although osteoclast stimulatory transmembrane protein (OC-STAMP) is implicated in Silicosis progression, its regulatory mechanisms are not fully understood. In this study, we detected elevated OC-STAMP expression in Silicosis patients and found that treatment with OC-STAMP siRNA can alleviate the progression of Silicosis in mice, suggesting the potential of OC-STAMP as a diagnostic and therapeutic target for Silicosis. METHODS First, rat models of Silicosis were developed at various stages. A suite of histological and molecular techniques, including Hematoxylin and eosin (HE), Masson, Prussian blue staining, and immunohistochemistry, along with real-time polymerase chain reaction (RT-PCR), were employed to assess the expression levels of OC-STAMP, as well as indicators of ferroptosis and fibrosis.Second, MLE-12 cells were cultured in vitro to establish an OC-STAMP overexpression model, and the relationship between OC-STAMP and ferroptosis was evaluated using flow cytometry, and western blotting. Subsequently, to verify the role of OC-STAMP and ferroptosis in Silicosis progression, we administered OC-STAMP siRNA and Fer-1 to Silicosis mice respectively. Whole-body volumetric plethysmography (WBP) was utilized to assess the respiratory function of the mice, and Micro-CT was applied to detect the lung nodules in the mice. The levels of OC-STAMP, as well as indexes associated with ferroptosis and fibrosis, were assessed using Hematoxylin and eosin (HE), Masson, Sirius red staining, immunohistochemistry, and western blot analysis. The polarization of macrophages towards M1 and M2 phenotypes in lung tissues was analyzed by flow cytometry. Ultimately, the plasma expression of OC-STAMP in patients diagnosed with Silicosis was quantified using enzyme-linked immunosorbent assay (ELISA). RESULTS In vivo experiments showed that OC-STAMP accelerates the fibrotic process of Silicosis, which may promote the epithelial-mesenchymal transformation by triggering ferroptosis of alveolar type II epithelial cells, and thus promote the progression of Silicosis. Furthermore, in vitro studies indicated that OC-STAMP overexpression causes ferroptosis in alveolar type II epithelial cells which contributes to fibrosis. Notably, treatment with siRNA in Silicosis mice confirmed that OC-STAMP inhibition effectively mitigates ferroptosis and retarded the progression of Silicosis fibrosis. Plasma of patients with Silicosis exhibited elevated OC-STAMP levels. CONCLUSIONS Overall, OC-STAMP induces ferroptosis and exacerbates fibrosis in Silicosis. OC-STAMP siRNA and Fer-1 mitigate abnormal collagen deposition and delay the progression of Silicosis. Collectively, these findings highlight the potential of OC-STAMP as a novel biomarker for diagnosing and treating Silicosis.
Collapse
Affiliation(s)
- Jing Wu
- Basic Medicine Department, Xiamen Medical College, Xiamen, 361023, Fujian, China
- Public Health School of Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Bingyu Zhang
- Basic Medicine Department, Xiamen Medical College, Xiamen, 361023, Fujian, China
- Public Health School of Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Wei Du
- Basic Medicine Department, Xiamen Medical College, Xiamen, 361023, Fujian, China
- Public Health School of Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Yimin Shi
- Basic Medicine Department, Xiamen Medical College, Xiamen, 361023, Fujian, China
| | - Chunhong Xie
- Department of Clinical Laboratory, the Second Affiliated Hospital of Xiamen Medical College, Xiamen, 361021, Fujian, China
| | - Yanyan Ke
- Basic Medicine Department, Xiamen Medical College, Xiamen, 361023, Fujian, China.
- Insitute of Respiratory Research, Xiamen Medical College, Xiamen, 361023, Fujian, China.
| | - Xue Yi
- Basic Medicine Department, Xiamen Medical College, Xiamen, 361023, Fujian, China.
- Public Health School of Fujian Medical University, Fuzhou, 350122, Fujian, China.
- Insitute of Respiratory Research, Xiamen Medical College, Xiamen, 361023, Fujian, China.
| |
Collapse
|
28
|
Farhadi R, Daniali M, Baeeri M, Khorasani R, Haghi-Aminjan H, Gholami M, Rahimifard M, Navaei-Nigjeh M, Abdollahi M. Molecular evidence of the inhibitory potential of melatonin against sodium arsenite toxicity. Heliyon 2025; 11:e42113. [PMID: 39916822 PMCID: PMC11799970 DOI: 10.1016/j.heliyon.2025.e42113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
Introduction Sodium arsenite (SA), NaAsO2, is among the most hazardous toxicants, and wide use and presence of this toxicant leads to a severe environmental threat. Exposure to SA is associated with many health concerns, such as the prevalence of cancer and diabetes mellitus type 2 (DMT2). Many studies suggest that SA induces inflammation and biochemical impairments through different mechanisms, including increasing oxidative stress and altering vital genes such as biochemical and anti-inflammatory. Recent studies on melatonin (MLT), a harmless hormone secreted in the body generally for induction of sleepiness, find many beneficial and positive effects. Mitigating different harms and toxicities through different mechanisms, such as antioxidant properties, anti-inflammatory effects, and critical gene regulation, is essential. Due to these findings, this study aimed to evaluate the hypothesis that MLT may ameliorate pancreatic damage caused by exposure to SA. Methods Forty-eight adult healthy male wistar rats aged 7-8 weeks were divided into eight for this research. Group 1 did not receive any intervention. Group 2 received 10 mg/kg/day MLT through intraperitoneal (IP) injection. Groups 3, 4, and 5 received 1.5 (1/10 LD50), 5 (1/3 LD50), and 7.5 (1/2 LD50) mg/kg SA, respectively. Groups 6, 7, and 8 were given 1.5 (1/10 LD50), 5 (1/3 LD50), and 7.5 (1/2 LD50) mg/kg of SA along with 10 mg/kg/day MLT, respectively, during the last ten days of the experiment. After 28 days of the experiment, the blood and tissue samples of the pancreas were removed for biochemical and pathological examination. Results MLT attenuates SA toxicity by reducing oxidative stress biomarkers and inflammation markers. Moreover, MLT improves SA exposure's biochemical and functional damages by regulating related genes and pathways. Conclusion MLT poses protective and preventive effects on the pancreas against exposure to SA. However, MLT's therapeutic and beneficial impacts have great potential for further investigation.
Collapse
Affiliation(s)
- Ramtin Farhadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Marzieh Daniali
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Maryam Baeeri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Reza Khorasani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hamed Haghi-Aminjan
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mahban Rahimifard
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mona Navaei-Nigjeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
29
|
Gholipour A, Zahedmehr A, Arabian M, Shakerian F, Maleki M, Oveisee M, Malakootian M. MiR-6721-5p as a natural regulator of Meta-VCL is upregulated in the serum of patients with coronary artery disease. Noncoding RNA Res 2025; 10:25-34. [PMID: 39296643 PMCID: PMC11406674 DOI: 10.1016/j.ncrna.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 09/21/2024] Open
Abstract
Background Coronary artery disease (CAD), the leading cause of mortality globally, arises from atherosclerotic blockage of the coronary arteries. Meta-vinculin (meta-VCL), a large spliced isoform of VCL, co-localizes in muscular adhesive structures and plays significant roles in cardiac physiology and pathophysiology. This study aimed to identify microRNAs (miRNAs) regulating meta-VCL expression and investigate the expression alterations of the miRNAs of interest and meta-VCL as potential biomarkers in the serum of CAD patients. Methods Bioinformatics tools were employed to select miRNAs targeting meta-VCL. Cell-based ectopic expression analysis and a dual-luciferase assay were used to examine the interactions between miRNAs and meta-VCL. An ELISA assessed the concentrations of interleukin-6 (IL-6), IL-10, and tumor necrosis factor-α (TNF-α). MiRNA and meta-VCL expression patterns and biomarker suitability were evaluated in serum samples from CAD and non-CAD individuals using real-time PCR. A cardiac cell-line data set and CAD blood exosome samples were analyzed using bioinformatics and ROC curve analyses, respectively. Results miR-6721-5p directly interacted with the putative target sites at the 3'-UTR of meta-VCL and regulated its expression. IL-10 and TNF-α concentrations, which may act as anti-inflammatory factors, decreased following miR-6721-5p upregulation and meta-VCL downregulation. Bioinformatics and experimental expression analyses confirmed downregulated meta-VCL expression and upregulated miR-6721-5p expression in CAD samples. ROC curve analysis yielded an AUC score of 0.705 (P = 0.018), indicating the potential suitability of miR-6721-5p as a biomarker for CAD. Conclusions miR-6721-5p plays a regulatory role in meta-VCL expression and may contribute to CAD development by reducing anti-inflammatory factors. These findings suggest that miR-6721-5p could serve as a novel biomarker in the pathogenesis of CAD.
Collapse
Affiliation(s)
- Akram Gholipour
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Zahedmehr
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maedeh Arabian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Farshad Shakerian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
- Cardiovascular Intervention Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Maleki
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mahshid Malakootian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Su Y, Huang M, Chen Q, He J, Li S, Wang M. Harnessing β-glucan conjugated quercetin nanocomplex to function as a promising anti-inflammatory agent via macrophage-targeted delivery. Carbohydr Polym 2025; 349:122952. [PMID: 39638531 DOI: 10.1016/j.carbpol.2024.122952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/11/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024]
Abstract
Quercetin, a promising anti-inflammatory agent, faces challenges related to poor bioavailability and limited practical applications. β-glucan, a natural polysaccharide, can be specifically recognized by macrophages, making it an ideal targeting carrier to enhance therapeutic efficacy for macrophage-related dysfunctions. In this study, β-glucan conjugated quercetin nano-complexes (CM-Cur@QT) were developed to target macrophage and alleviate pro-inflammatory response in M1-like macrophages. The results demonstrated that CM-Cur@QT exhibited a spheric shape with an average diameter around 200 nm. FT-IR, 1H NMR, XRD and XPS analyses confirmed the complexation of CM-Cur@QT. This complex showed excellent stability during stimulated digestion, protecting QT from degradation while maintaining favorable antioxidant activity. After complexation, CM-Cur@QT displayed sustained uptake kinetics and enhanced accumulation in macrophages, with a 61.88 % increase compared to individual quercetin after 5 h of incubation. Meanwhile, CM-Cur@QT administration induced evidently cell cycle phases transitions and altered phagocytotic activity in M1-like macrophages. Furthermore, CM-Cur@QT reduced intracellular ROS accumulation, achieving a ROS scavenging rate of up to 49.92 %, compared to 25.59 % in quercetin group. This complex also effectively modulated TNF-a, IL-6 and TGF-β secretion profiles in pro-inflammatory macrophages, outperforming individual QT treatment. Notably, CM-Cur@QT facilitated anti-inflammatory effects while minimizing impacts on inactivated M0 macrophages. These findings underscore the potential of CM-Cur@QT as a promising agent for mitigating inflammatory disorders.
Collapse
Affiliation(s)
- Yuting Su
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China
| | - Manting Huang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China
| | - Qiaochun Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China; Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China
| | - Jiayi He
- Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China; College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Siqian Li
- Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China; College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China
| | - Mingfu Wang
- Shenzhen Key Laboratory of Food Nutrition and Health, Shenzhen University, Shenzhen 518060, China; College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
31
|
Engel K, Kulow VA, Chernyakov D, Willscher E, Fähling M, Edemir B. Segment specific loss of NFAT5 function in the kidneys is sufficient to induce a global kidney injury like phenotype. FASEB J 2025; 39:e70352. [PMID: 39874047 PMCID: PMC11774485 DOI: 10.1096/fj.202402497r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/17/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Nuclear factor of activated T-cells 5 (NFAT5) is a transcription factor known for its role in osmotic stress adaptation in the renal inner medulla, due to the osmotic gradient that is generated between the renal cortex and renal inner medulla. However, its broader implications in kidney injury and chronic kidney disease (CKD) are less understood. Here we used two different Cre deleter mice (Ksp1.3-Cre and Aqp2-Cre) to generate tubule segment and even cell type-specific NFAT5-deficient mice and performed extensive gene expression profiling. In both Nfat5 knockout models, we observed massive changes in gene expression pattern, with heightened inflammatory responses and renal injury, culminating in renal fibrosis. Interestingly, inflammatory responses were much more prominent in the Aqp2Cre+/-Nfat5fl/fl mice that lack NFAT5 only in the collecting duct. By analyzing gene expression in the medullary and cortical regions of the kidney separately, we confirmed that the loss of NFAT5 results in kidney injury that extends beyond hypertonic areas. Renal injury correlates with the expression level of genes involved in inflammatory response, injury severity, and cytokine signaling. Thus, NFAT5 is essential not only for adapting to osmotic stress but also for its loss of function, which induces activation of inflammatory response and cytokine signaling that might affect regions with functional NFAT5 expression.
Collapse
Affiliation(s)
- Kristina Engel
- Department of Medicine, Hematology and OncologyMartin Luther University Halle‐WittenbergHalle (Saale)Germany
| | - Vera Anna Kulow
- Institute of Translational PhysiologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Dmitry Chernyakov
- Department of Medicine, Hematology and OncologyMartin Luther University Halle‐WittenbergHalle (Saale)Germany
| | - Edith Willscher
- Department of Medicine, Hematology and OncologyMartin Luther University Halle‐WittenbergHalle (Saale)Germany
| | - Michael Fähling
- Institute of Translational PhysiologyCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
| | - Bayram Edemir
- Department of Medicine, Hematology and OncologyMartin Luther University Halle‐WittenbergHalle (Saale)Germany
- Institute for Physiology and Pathophysiology, Zentrum für Biomedizinische Ausbildung und Forschung (ZBAF)Witten/Herdecke UniversityWittenGermany
| |
Collapse
|
32
|
Guo K, Joshipura K, Ricart K, Patel RP, Gower BA, Andriankaja OM, Morou-Bermudez E. Association of over-the-counter mouthwash use with markers of nitric oxide metabolism, inflammation, and endothelial function-a cross-sectional study. FRONTIERS IN ORAL HEALTH 2025; 6:1488286. [PMID: 39981124 PMCID: PMC11841417 DOI: 10.3389/froh.2025.1488286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/07/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Regular use of mouthwash can disrupt nitrate reduction by oral bacteria and may affect systemic nitric oxide (NO) levels, which are important for inflammation and endothelial function. We aim to assess the association between over-the-counter (OTC) mouthwash use and nitrate/nitrite, markers of inflammation (IL-6, TNF-α, CRP) and endothelial function (sICAM-1, sVCAM-1) in serum and saliva, and to assess the relationship between nitrate/nitrite levels and these biomarkers, as well as how OTC mouthwash modulated this relationship. We hypothesize that nitrates/nitrites are associated with these biomarkers, and that their associations would vary with the frequency of mouthwash use. Method Our cross-sectional study used data and specimen from the baseline of the San Juan Overweight Adult Longitudinal Study (SOALS). Robust Gamma regression with log-link function, Spearman correlations and partial correlations adjusted for covariates were used for the analysis. Results Using OTC mouthwash twice a day or more was significantly associated with lower serum nitrite levels compared to less frequent use (β = -0.357, 95% CI: -0.650, -0.064), but not with other markers of inflammation and endothelial function. Mouthwash use differentially impacted the relationship between nitrate/nitrite and TNF-α, sICAM-1 and sVCAM-1. Specifically, in the participants who used mouthwash less than twice a day or no use, TNF-α (β = -0.35, 95% CI: -0.52, -0.18), and sICAM-1 (β = -0.21, 95% CI: -0.32, -0.09) were negatively associated with serum nitrite. In the participants who used mouthwash twice a day or more use, TNF-α was positively associated with serum nitrate (β = 3.36, 95% CI: 2.07, 4.65), salivary nitrite (β = 1.04, 95% CI: 0.39, 1.69) and salivary nitrate (β = 0.48, 95% CI: 0.25, 0.71); sICAM-1 was positively associated with serum nitrate (β = 1.58, 95% CI: 0.86, 2.29). In both subgroups of mouthwash users, sVCAM-1 was positively correlated with serum nitrate and salivary nitrate. In addition, sVCAM-1 was positively correlated with serum nitrite in participants who used mouthwash frequently (ρ_S = 0.18, p = 0.045). Discussion Regular use of OTC mouthwash was associated with systemic nitric oxide. This raises concerns about its potential effects on the levels of inflammatory and endothelial biomarkers associated with cardiometabolic diseases.
Collapse
Affiliation(s)
- Kai Guo
- Surgical Science Department, School of Dental Medicine, University of Puerto Rico, San Juan, PR, United States
| | | | - Karina Ricart
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rakesh P. Patel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Barbara A. Gower
- Department of Nutrition Sciences, Division of Physiology & Metabolism, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Oelisoa Mireille Andriankaja
- Center for Oral Health Research (COHR), College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - Evangelia Morou-Bermudez
- Surgical Science Department, School of Dental Medicine, University of Puerto Rico, San Juan, PR, United States
| |
Collapse
|
33
|
Carroll-Portillo A, Barnes O, Coffman CN, Braun CA, Singh SB, Lin HC. Transcytosis of T4 Bacteriophage Through Intestinal Cells Enhances Its Immune Activation. Viruses 2025; 17:134. [PMID: 39861923 PMCID: PMC11769353 DOI: 10.3390/v17010134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Interactions between bacteriophages with mammalian immune cells are of great interest and most phages possess at least one molecular pattern (nucleic acid, sugar residue, or protein structure) that is recognizable to the immune system through pathogen associated molecular pattern (PAMP) receptors (i.e., TLRs). Given that phages reside in the same body niches as bacteria, they share the propensity to stimulate or quench immune responses depending on the nature of their interactions with host immune cells. While most in vitro research focuses on the outcomes of direct application of phages to immune cells of interest, the potential impact of their transcytosis through the intestinal barrier has yet to be considered. As transcytosis through intestinal cells is a necessary step in healthy systems for access by phage to the underlying immune cell populations, it is imperative to understand how this step may play a role in immune activation. We compared the activation of macrophages (as measured by TNFα secretion) following direct phage application to those stimulated by incubation with phage transcytosed through a polarized Caco2 epithelial barrier model. Our results demonstrate that phages capable of activating TNFα secretion upon direct contact maintain the stimulatory capability following transcytosis. Furthermore, activation of macrophages by a transcytosed phage is enhanced as compared to that occurring with an equivalent multiplicity of directly applied phage.
Collapse
Affiliation(s)
- Amanda Carroll-Portillo
- Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM 87131, USA
| | - October Barnes
- Biomedical Research Institute of New Mexico, Albuquerque, NM 87108, USA; (O.B.); (C.N.C.); (C.A.B.); (S.B.S.)
| | - Cristina N. Coffman
- Biomedical Research Institute of New Mexico, Albuquerque, NM 87108, USA; (O.B.); (C.N.C.); (C.A.B.); (S.B.S.)
| | - Cody A. Braun
- Biomedical Research Institute of New Mexico, Albuquerque, NM 87108, USA; (O.B.); (C.N.C.); (C.A.B.); (S.B.S.)
| | - Sudha B. Singh
- Biomedical Research Institute of New Mexico, Albuquerque, NM 87108, USA; (O.B.); (C.N.C.); (C.A.B.); (S.B.S.)
| | - Henry C. Lin
- Division of Gastroenterology and Hepatology, University of New Mexico, Albuquerque, NM 87131, USA
- Medicine Service, New Mexico VA Health Care System, Albuquerque, NM 87108, USA
| |
Collapse
|
34
|
Rao SP, Imam-Fulani AO, Xie W, Phillip S, Chennavajula K, Lind EB, Zhang Y, Vince R, Lee MK, More SS. Oral prodrug of a novel glutathione surrogate reverses metabolic dysregulation and attenuates neurodegenerative process in APP/PS1 mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633247. [PMID: 39868172 PMCID: PMC11761491 DOI: 10.1101/2025.01.15.633247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Glycation-induced oxidative stress underlies the numerous metabolic ravages of Alzheimer's disease (AD). Reduced glutathione levels in AD lead to increased oxidative stress, including glycation-induced pathology. Previously, we showed that the accumulation of reactive 1,2-dicarbonyls such as methylglyoxal, the major precursor of non-enzymatic glycation products, was reduced by the increased function of GSH-dependent glyoxalase-1 enzyme in the brain. In this two-pronged study, we evaluate the therapeutic efficacy of an orally bioavailable prodrug of our lead glyoxalase substrate, pro-ψ-GSH, for the first time in a transgenic Alzheimer's disease mouse model. This prodrug delivers pharmacodynamically relevant brain concentrations of ψ-GSH upon oral delivery. Chronic oral dosing of pro-ψ-GSH effectively reverses the cognitive decline observed in the APP/PS1 mouse model. The prodrug successfully mirrors the robust effects of the parent drug i.e., reducing amyloid pathology, glycation stress, neuroinflammation, and the resultant neurodegeneration in these mice. We also report the first metabolomics study of such a treatment, which yields key biomarkers linked to the reversal of AD-related metabolic dysregulation. Collectively, this study establishes pro-ψ-GSH as a viable, disease-modifying therapy for AD and paves the way for further preclinical advancement of such therapeutics. Metabolomic signatures identified could prove beneficial in the development of treatment-specific clinically translatable biomarkers.
Collapse
Affiliation(s)
- Swetha Pavani Rao
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Aminat O. Imam-Fulani
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Wei Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Samuel Phillip
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Krishna Chennavajula
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Erin B. Lind
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ying Zhang
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, USA
| | - Robert Vince
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Michael K. Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Swati S. More
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
35
|
Palmieri TL, Heard J. Biomarkers of sepsis in burn injury: an update. BURNS & TRAUMA 2025; 13:tkae080. [PMID: 39822649 PMCID: PMC11736899 DOI: 10.1093/burnst/tkae080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/16/2024] [Indexed: 01/19/2025]
Abstract
Sepsis, a dysregulated response to infection, is a leading cause of death after burn injury. Changes in the immune response as well as the loss of the skin, the primary barrier to infection, contribute to the increased risk for infection and sepsis in burn patients. This higher risk is further compounded by the development of the systemic inflammatory response and hypermetabolic state, which limit the utility of commonly used infection markers. As such, the development of sepsis biomarkers after burn injury is an imperative. A sepsis biomarker would facilitate earlier diagnosis and treatment of sepsis, thus decreasing length of stay, morbidity, and mortality after burn injury. Numerous different biomarkers, ranging from acute phase reactants, cytokines, and inflammatory markers to omics analyses and extracellular vesicles have been assessed as potential biomarkers in burn sepsis. To date no single biomarker has proven useful as the sole indicator for sepsis. The future of burn sepsis biomarkers will likely require a panel of biomarkers from all categories. The purpose of this review article is to list the various biomarkers that have been studied in burn sepsis and describe their clinical utility and future use in patients with burn injury.
Collapse
Affiliation(s)
- Tina L Palmieri
- University of California Davis, Shriners Children’s Northern California, Burn Division, Department of Surgery, University of California, Davis, 2335 Stockton Blvd, Sacramento, CA 95817, United States
| | - Jason Heard
- University of California Davis, Shriners Children’s Northern California, Burn Division, Department of Surgery, University of California, Davis, 2335 Stockton Blvd, Sacramento, CA 95817, United States
| |
Collapse
|
36
|
Wang J, Ma L, Fang Y, Ye T, Li H, Lan P. Factors influencing glycocalyx degradation: a narrative review. Front Immunol 2025; 15:1490395. [PMID: 39885987 PMCID: PMC11779607 DOI: 10.3389/fimmu.2024.1490395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/30/2024] [Indexed: 02/01/2025] Open
Abstract
The glycocalyx is a layer of villus-like structure covering the luminal surface of vascular endothelial cells. Damage to the glycocalyx has been proven linked to the development of many diseases. However, the factors that promote damage to the glycocalyx are not fully elaborated. This review summarizes factors leading to the reduction of the glycocalyx in detail, including inflammatory factors, ischemia-reperfusion, oxidative stress, lipids, glucose, high sodium, female sex hormones and others. Additionally, the mechanisms underlying its degradation are discussed. To better prevent and treat related diseases induced by glycocalyx degradation, it is a meaningful measure to avoid these factors.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cardiology, Nanning Hospital of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Lan Ma
- Department of Neurology, Wenzhou Traditional Chinese Medicine (TCM) Hospital of Zhejiang Chinese Medical University, Wenzhou, Zhejiang, China
| | - Yu Fang
- Department of Cardiology, Nanning Hospital of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Tengteng Ye
- Department of Cardiology, Nanning Hospital of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Hongbo Li
- Department of Cardiology, Nanning Hospital of Traditional Chinese Medicine, Nanning, Guangxi, China
| | - Peng Lan
- Department of Cardiology, Nanning Hospital of Traditional Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
37
|
Dokumacioglu E, Iskender H, Kapakin KAT, Bolat İS, Mokhtare B, Dogan Omur A, Hayirli A. Curcumin exerts protective effects against valproic acid-induced testicular damage through modulating the JAK1/STAT-3/IL-6 signaling pathway in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:230-236. [PMID: 39850114 PMCID: PMC11756740 DOI: 10.22038/ijbms.2024.76948.16659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 09/15/2024] [Indexed: 01/25/2025]
Abstract
OBJECTIVES This experiment was carried out to investigate the protective effects of curcumin (CUR) on testicular damage induced by the valproic acid (VPA) administration. MATERIALS AND METHODS Male Wistar-Albino rats (n=28, 250-300 g) were randomly divided into four groups: Control (1 ml saline, oral), VPA (500 mg/kg, IP), CUR (200 mg/kg, oral), or VPA+CUR (500 mg/kg, VPA, IP plus 200 mg/kg CUR, oral). The treatments were applied for 14 days. Serum testosterone and testis [Janus kinases1 (JAK1), signal transducers and activators of transcription-3 (STAT-3), interleukin-6 (IL-6), malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-α), interleukin-18 (IL-18), and nuclear factor (NF)-κB)] samples were collected for biochemical analyses. Semen samples were subjected to microscopy for spermatological parameters. Testis tissue was also analyzed for histopathological and immunohistochemical methods. RESULTS The VPA administration caused a 37% decrease in serum testosterone concentration and 5.32, 9.51, 2.44, and 3.68-fold increases in testicular tissue JAK1, STAT-3, IL-6, and MDA levels, respectively. There were also 50, 52, and 72% reductions in sperm motility, sperm viability, and the mean testicular biopsy score, respectively, accompanied by considerable degenerative changes and necrosis in seminiferous tubules in the VPA group. There is also an immune-positive reaction for IL-18 and NF-κB in only Leydig cells. CONCLUSION The CUR treatment may be beneficial in restoring testicular damage through antiinflammatory and anti-oxidant potential.
Collapse
Affiliation(s)
- Eda Dokumacioglu
- Eda Dokumacioglu, Department of Nutrition and Dietetics, Faculty of Health Sciences, Artvin Coruh University, Artvin 08000, Turkey
| | - Hatice Iskender
- Eda Dokumacioglu, Department of Nutrition and Dietetics, Faculty of Health Sciences, Artvin Coruh University, Artvin 08000, Turkey
| | | | - İSmail Bolat
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum 25240, Turkey
| | - Behzat Mokhtare
- Department of Pathology, Faculty of Veterinary Medicine, Dicle University, Diyarbakır, 21000, Turkey
| | - Ali Dogan Omur
- Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, Ataturk University, Erzurum 25240, Turkey
| | - Armagan Hayirli
- Department of Animal Nutrition and Nutritional Disorders, Faculty of Veterinary Medicine, Ataturk University, Erzurum 25240, Turkey
| |
Collapse
|
38
|
Kalogriopoulos NA, Tei R, Yan Y, Klein PM, Ravalin M, Cai B, Soltesz I, Li Y, Ting AY. Synthetic GPCRs for programmable sensing and control of cell behaviour. Nature 2025; 637:230-239. [PMID: 39633047 PMCID: PMC11666456 DOI: 10.1038/s41586-024-08282-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024]
Abstract
Synthetic receptors that mediate antigen-dependent cell responses are transforming therapeutics, drug discovery and basic research1,2. However, established technologies such as chimeric antigen receptors3 can only detect immobilized antigens, have limited output scope and lack built-in drug control3-7. Here we engineer synthetic G-protein-coupled receptors (GPCRs) that are capable of driving a wide range of native or non-native cellular processes in response to a user-defined antigen. We achieve modular antigen gating by engineering and fusing a conditional auto-inhibitory domain onto GPCR scaffolds. Antigen binding to a fused nanobody relieves auto-inhibition and enables receptor activation by drug, thus generating programmable antigen-gated G-protein-coupled engineered receptors (PAGERs). We create PAGERs that are responsive to more than a dozen biologically and therapeutically important soluble and cell-surface antigens in a single step from corresponding nanobody binders. Different PAGER scaffolds allow antigen binding to drive transgene expression, real-time fluorescence or endogenous G-protein activation, enabling control of diverse cellular functions. We demonstrate multiple applications of PAGER, including induction of T cell migration along a soluble antigen gradient, control of macrophage differentiation, secretion of therapeutic antibodies and inhibition of neuronal activity in mouse brain slices. Owing to its modular design and generalizability, we expect PAGERs to have broad utility in discovery and translational science.
Collapse
Affiliation(s)
| | - Reika Tei
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Yuqi Yan
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Peter M Klein
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Matthew Ravalin
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Bo Cai
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Yulong Li
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, Beijing, China
| | - Alice Y Ting
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Department of Biology, Stanford University, Stanford, CA, USA.
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA, USA.
- Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
39
|
Monaghan NP, Shah S, Keith BA, Nguyen SA, Newton DA, Baatz JE, Wagner CL, Rizk HG. Proinflammatory Cytokine Profiles in Menière's Disease and Vestibular Migraine. Otol Neurotol 2025; 46:88-95. [PMID: 39627868 DOI: 10.1097/mao.0000000000004372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
OBJECTIVE To evaluate the levels of inflammatory cytokines and symptom survey scores in patients diagnosed with Menière's disease or vestibular migraine from a single center by a single neurotologist compared to control subjects with no history of dizziness or migraine. STUDY DESIGN Cross-sectional pilot study. SETTING Single-center tertiary referral center in Charleston, SC. PATIENTS Patients were recruited from the neurotology clinic at a tertiary referral center. Patients with definite Menière's disease or definite vestibular migraine as defined by the Barany consensus criteria were eligible. Control subjects presented to clinic without dizziness, vertigo, fluctuating hearing loss, or a history of migraine. MAIN OUTCOME MEASURES Questionnaire scores include DHI, SF-20, CFQ, PHQ-9, PSWQ, GAD-7, NVI, DCS, VM-PATHI, and MD-POSI. Circulating and in vitro levels of cytokines include ENA-78, GROα, IFN-α2a, IFN-γ, IL-10, IL-1α, IL-1β, IL-1RA, IL-2, IL-4, IL-5, IL-6, IL-8, MCP-1, MCP-2, MDC, MIP-1α, MIP-1β, and TNF-α. Cytokine levels were compared with effect size analysis. RESULTS There were 20 Menière's disease, 20 vestibular migraine, and 10 control patients enrolled in this study. Episode frequencies ranged from three per week to two to five per year in the MD group and daily to 1 every 2 to 3 months in the VM group. When patient-derived PBMC samples were compared to vestibular migraine, TNF-α ( d = -0.427 [-0.879, 0.025]) and IFN-γ ( d = -0.818 [-1.313, -0.323]) were found to be higher in Menière's disease, whereas ENA-78 ( d = -0.652 [-1.361, 0.056]) was found to be lower. No differences were found when cytokines were measured following stimulation with LPS. CONCLUSIONS This pilot study suggests Menière's disease patients may have higher levels of TNF-α and IFN-γ and lower levels of ENA-78 than vestibular migraine patients when measured following in vitro release from patient-derived PBMC. Increased sample size, optimized blood draw timing, and more specific PBMC stimulation may help us further elucidate inflammatory pathways implicated in these disorders.
Collapse
Affiliation(s)
| | | | | | | | - Danforth A Newton
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - John E Baatz
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Carol L Wagner
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Habib G Rizk
- Department of Otolaryngology-Head and Neck Surgery
| |
Collapse
|
40
|
Mahmoudi A, Butler AE, Orekhov AN, Jamialahmadi T, Sahebkar A. Statins as a Potential Treatment for Non-alcoholic Fatty Liver Disease: Target Deconvolution using Protein-protein Interaction Network Analysis. Curr Med Chem 2025; 32:1355-1377. [PMID: 37644746 DOI: 10.2174/0929867331666230829164832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/28/2023] [Accepted: 07/18/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND The hallmark of non-alcoholic fatty liver disease (NAFLD) is aberrant buildup of triglycerides (TGs) in hepatocytes. Many genes promote NAFLD development. Using bioinformatics tools, we investigated the possible effect of statins on genes involved in NAFLD progression. METHODS Protein interactions of statins and NAFLD were searched in gene-drug and gene-disease databases. A Protein-Protein interaction (PPI) network was constructed to find hub genes and Molecular Complex Detection (MCODE) of NAFLD-related genes. Shared protein targets between protein targets of statins and NAFLD-associated genes were identified. Next, targets of each statin were assayed with all modular clusters in the MCODEs related to NAFLD. Biological process and pathway enrichment analysis for shared proteins was performed. RESULTS Screening protein targets for conventional statins and curated NAFLD-related genes identified 343 protein targets and 70 genes, respectively. A Venn diagram of NAFLD-related genes and protein targets of statins showed 24 shared proteins. The biological pathways on KEGG enrichment associated with the 24 shared protein sets were evaluated and included cytokine-cytokine receptor interaction, adipocytokine, PPAR, TNF and AMPK signaling pathways. Gene Ontology analysis showed major involvement in lipid metabolic process regulation and inflammatory response. PPI network analysis of 70 protein targets indicated 13 hub genes (PPARA, IL4, CAT, LEP, SREBF1, PRKCA, CYP2E1, NFE2L2, PTEN, NR1H4, ADIPOQ, GSTP1 and TGFB1). Comparing all seven statins with the three MCODE clusterings and 13 hub genes revealed that simvastatin as the most associated statin with NAFLD. CONCLUSION Simvastatin has the most impact on NAFLD-related genes versus other statins.
Collapse
Affiliation(s)
- Ali Mahmoudi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Department of Medical Sciences, Royal College of Surgeons in Ireland Bahrain, Adliya, Bahrain
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow 125315, Russia
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, Moscow 121609, Russia
| | - Tannaz Jamialahmadi
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
41
|
Thangadurai M, Sethuraman S, Subramanian A. Drug Delivery Approaches for Rheumatoid Arthritis: Recent Advances and Clinical Translation Aspects. Crit Rev Ther Drug Carrier Syst 2025; 42:1-54. [PMID: 40084516 DOI: 10.1615/critrevtherdrugcarriersyst.v42.i3.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Rheumatoid arthritis (RA) is a multifactorial autoimmune disease characterized with symmetrical progression of joint deformity that is often diagnosed at a chronic condition with other associated pathological conditions such as pericarditis, keratitis, pulmonary granuloma. Despite the understanding of RA pathophysiology in disease progression, current clinical treatment options such as disease-modifying anti-rheumatic drugs (DMARDs), biologics, steroids, and non-steroidal anti-inflammatory drugs (NSAIDs) provide only palliative therapy while causing adverse side effects such as off-target multi-organ toxicity and risk of infections. Further, available drug delivery strategies to treat RA pathogenicity does not successfully reach the site of action due to various barriers such as phagocytosis and first pass effect in addition to the disease complexity and unknown etiology, thereby leading to the development of irreversible joint dysfunction. Therefore, novel and effective strategies remain an unmet need to control the disease progression and to maintain the balance between pro- and anti-inflammatory cytokines. This review provides a comprehensive outlook on the RA pathophysiology and its corresponding disease progression. Contributions of synoviocytes such as macrophages, fibroblast-like cells in increasing invasiveness to exacerbate joint damage is also outlined in this review, which could be a potential future therapeutic target to complement the existing treatment regimens in controlling RA pathogenesis. Further, various smart drug delivery approaches under research to achieve maximum therapeutic efficacy with minimal adverse side effects have been discussed, which in turn emphasize the unmet challenges and future perspectives in addressing RA complications.
Collapse
Affiliation(s)
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Laboratory, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| | - Anuradha Subramanian
- Tissue Engineering & Additive Manufacturing (TEAM) Laboratory, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| |
Collapse
|
42
|
Wang B, Hasturk O, Kumarasinghe U, Rudolph S, Staii C, Chen Y, Kaplan DL. Temporary Nanoencapsulation of Human Intestinal Organoids Using Silk Ionomers. Adv Healthc Mater 2025; 14:e2403176. [PMID: 39648539 DOI: 10.1002/adhm.202403176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/03/2024] [Indexed: 12/10/2024]
Abstract
Human intestinal organoids (HIOs) are vital for modeling intestinal development, disease, and therapeutic tissue regeneration. However, their susceptibility to stress, immunological attack, and environmental fluctuations limits their utility in research and therapeutic applications. This study evaluated the effectiveness of temporary silk protein-based layer-by-layer (LbL) nanoencapsulation technique to enhance the viability and functions of HIOs against common biomedical stressors, without compromising their native functions. Cell viability and differentiation capacity are assessed, finding that nanoencapsulation significantly improved HIO survival under the various environmental perturbations studied without compromising cellular functionality. Post-stress exposures, the encapsulated HIOs still successfully differentiated into essential intestinal cell types such as enterocytes, goblet cells, enteroendocrine cells, and Paneth cells. Moreover, the silk nanocoatings effectively protected against environmental stressors such as ultraviolet (UV) light exposure, protease degradation, antibody binding, and cytokine-induced inflammation. This nanoencapsulation technique shows promise for advancing HIO applications in disease modeling, drug testing, and potential transplantation therapies.
Collapse
Affiliation(s)
- Brooke Wang
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Onur Hasturk
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | | | - Sara Rudolph
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Cristian Staii
- Department of Physics and Astronomy, Tufts University, Medford, MA, USA
| | - Ying Chen
- Department of Physics and Astronomy, Tufts University, Medford, MA, USA
| | - David L Kaplan
- Department of Physics and Astronomy, Tufts University, Medford, MA, USA
| |
Collapse
|
43
|
Illingworth EJ, Rychlik KA, Maertens A, Sillé FCM. Sex-specific transcriptomic effects of low-dose inorganic arsenic exposure on bone marrow-derived macrophages. Toxicology 2025; 510:153988. [PMID: 39515575 PMCID: PMC12023008 DOI: 10.1016/j.tox.2024.153988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/19/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Both tissue-resident macrophages and monocytes recruited from the bone marrow that transform into tissue-resident cells play critical roles in mediating homeostasis as well as in the pathology of inflammatory diseases. Inorganic arsenic (iAs) is the most common drinking water contaminant worldwide and represents a major public health concern. There are numerous diseases caused by iAs exposure in which macrophages are involved, including cardiovascular disease, cancer, and increased risk of (respiratory) infectious diseases. Notably, prenatal iAs exposure is also associated with negative birth outcomes and developmental immunotoxicity (DIT) contributing to long-term adverse outcomes of these immune-related diseases. Therefore, understanding the effects of iAs exposure on macrophages, particularly during immune development or tissue injury and inflammation, can help us better grasp the full range of arsenic immunotoxicity and better design therapeutic targets for iAs-induced diseases particularly in exposed populations. In contrast to prior published studies which often only focused on the effect of iAs on mature macrophages after development, in this study, we analyzed the transcriptome of M0-, M1- and M2-polarized male and female murine bone marrow-derived macrophages (BMDMs) which were exposed to iAs during the differentiation phase, as a model to study iAs (developmental) immunotoxicity. We identified differentially expressed genes by iAs in a sex- and stimulation-dependent manner and used bioinformatics tools to predict protein-protein interactions, transcriptional regulatory networks, and associated biological processes. Overall, our data suggest that M1-stimulated, especially female-derived, BMDMs are most susceptible to iAs exposure during differentiation. Most notably, we observed significant downregulation of major proinflammatory transcription factors, like IRF8, and its downstream targets, as well as genes encoding proteins involved in pattern recognition and antigen presentation, such as TLR7, TLR8, and H2-D1, potentially providing causal insight regarding the role of (early-life) arsenic exposure in perturbing immune responses to infectious diseases. We also observed significant downregulation of genes involved in processes crucial to coordinating a proinflammatory response including leukocyte migration, differentiation, and cytokine and chemokine production and response. Finally, we discovered that 24 X-linked genes were dysregulated in iAs-exposed female stimulation groups compared to only 3 across the iAs-exposed male stimulation groups. These findings elucidate the potential mechanisms underlying the sex-differential iAs-associated immune-related disease risk.
Collapse
Affiliation(s)
- Emily J Illingworth
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Kristal A Rychlik
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Public Health Program, School of Health Professions, Mayborn College of Health Sciences, University of Mary Hardin-Baylor, Belton, TX, USA
| | - Alexandra Maertens
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Fenna C M Sillé
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
44
|
Le TN, Bright R, Truong V, Li J, Juneja R, Vasilev K. Key biomarkers in type 2 diabetes patients: A systematic review. Diabetes Obes Metab 2025; 27:7-22. [PMID: 39355932 PMCID: PMC11618249 DOI: 10.1111/dom.15991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 10/03/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is not just a local health issue but a significant global health burden, affecting patient outcomes and clinical management worldwide. Despite the wealth of studies reporting T2DM biomarkers, there is an urgent need for a comparative review. This review aims to provide a comprehensive analysis based on the reported T2DM biomarkers and how these are linked with other conditions, such as inflammation and wound healing. A comparative review was conducted on 24 001 study participants, including 10 024 T2DM patients and 13 977 controls (CTL; age 30-90 years). Four main profiles were extracted and analysed from the clinical reports over the past 11 years: haematological (1084 cases vs. 1458 CTL), protein (6753 cases vs. 9613 CTL), cytokine (975 cases vs. 1350 CTL) and lipid (1212 cases vs. 1556 CTL). This review provides a detailed analysis of the haematological profile in T2DM patients, highlighting fundamental changes such as increased white blood cells and platelet counts, accompanied by decreases in red blood cell counts and iron absorption. In the serum protein profile, a reduction in albumin and anti-inflammatory cytokines was noted along with an increase in globulin levels and pro-inflammatory cytokines. Furthermore, changes in lipid profiles were discussed, specifically the decreases in high-density lipoprotein (HDL) and the increases in low-density lipoprotein (LDL) and triglycerides. Understanding the changes in these four biomarker profiles is essential for developing innovative strategies to create diagnostic and prognostic tools for diabetes management.
Collapse
Affiliation(s)
- Thien Ngoc Le
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Richard Bright
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Vi‐Khanh Truong
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Jordan Li
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
- Department of Renal Medicine, Flinders Medical CentreBedford ParkSouth AustraliaAustralia
| | - Rajiv Juneja
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
- Department of Renal Medicine, Flinders Medical CentreBedford ParkSouth AustraliaAustralia
| | - Krasimir Vasilev
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| |
Collapse
|
45
|
Riaz M, Rasool G, Yousaf R, Fatima H, Munir N, Ejaz H. Anti-Rheumatic potential of biological DMARDS and protagonistic role of bio-markers in early detection and management of rheumatoid arthritis. Innate Immun 2025; 31:17534259251324820. [PMID: 40091354 PMCID: PMC11912179 DOI: 10.1177/17534259251324820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease that primarily affects the synovial joint linings, resulting in progressive disability, increased mortality, and considerable economic costs. Early treatment with disease-modifying antirheumatic medications (DMARDs) can significantly improve the overall outlook for people with RA. Contemporary pharmaceutical interventions, encompassing standard, biological, and emerging small molecule disease- modifying anti-rheumatic medications continue to be the cornerstone of RA management, with substantial advancements made in the pursuit of achieving remission from the disease and preventing joint deformities. Nevertheless, a substantial segment of individuals with RA do not experience a satisfactory response to existing treatments, underscoring the pressing need for novel therapeutic options. Biologic DMARDs are among the therapy choices. Non-tumor necrosis factor inhibitors (Non-TNFi) such as abatacept, rituximab, tocilizumab, and sarilumab are examples, as are anti-tumor necrosis factor (TNF) medications such as infliximab, adalimumab, etanercept, golimumab, and certolizumab pegol. More recent biomarkers have emerged and showed usefulness in the early detection of RA. These biomarkers, often referred to simply as "biomarkers", are quantifiable indicators of normal or pathologic processes, and they can also gauge treatment response. The assessment of RA treatment response typically combines patient-reported outcomes, physical evaluations, and laboratory findings, as there isn't a single biomarker that has proven sufficient for measuring disease activity. This review explores the usage of biologic DMARDs as a therapeutic approach for RA, as well as the biomarkers typically used for RA early diagnosis, prognosis prediction, and disease activity evaluation.
Collapse
Affiliation(s)
- Muhammad Riaz
- Department of Allied Health Sciences, University of Sargodha, Sargodha, Pakistan
| | - Ghulam Rasool
- Department of Allied Health Sciences, University of Sargodha, Sargodha, Pakistan
| | - Ruhamah Yousaf
- Department of Health Professional Technologies, The University of Lahore, Lahore, Pakistan
| | - Hina Fatima
- Department of Biochemistry, Government College Women University, Faisalabad, Pakistan
| | - Naveed Munir
- Department of Biomedical Lab Sciences, School of Health Sciences, University of Management and Technology, Lahore, Pakistan
| | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| |
Collapse
|
46
|
Alhaddad R, Abualsoud BM, Al-Deeb I, Nsairat H. Green synthesized Zingiber officinale-ZnO nanoparticles: anticancer efficacy against 3D breast cancer model. Future Sci OA 2024; 10:2419806. [PMID: 39539163 PMCID: PMC11572278 DOI: 10.1080/20565623.2024.2419806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Aim: ZnO NPs were prepared via green synthesis utilizing Zingiber Officinale.Methodology: Physical characterization and biological activity were performed against 2D, and 3D spheroids MCF-7 cell lines.Results: The NPs exhibited 188.9, 175.7 and 171.2 nm size with charge of -8.2, -11.7 and -9.7 mV for the 2%, 3% and 4% formulations. XRD confirmed a wurtzite hexagonal phase. FTIR spectra showed Zn-O stretching vibrations. The 2%, 3% and 4% formulations presented IC50 values of 14.7, 26.2 and 47 μg/ml, respectively, with complete destruction of MCF-7 spheroids. Elevated TNF-α levels suggested an inflammatory-mediated mechanism of action.Conclusion: 2% Zingiber officinale-derived ZnO NPs showed antitumor potential against deserving further mechanistic and in vivo explorations.
Collapse
Affiliation(s)
- Ruqaya Alhaddad
- Pharmacological & Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Bassam M Abualsoud
- Pharmacological & Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Ibrahim Al-Deeb
- Pharmacological & Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
- Department of Clinical Pharmacy, Faculty of Pharmacy, Zarqa University, Zarqa, 13110, Jordan
| | - Hamdi Nsairat
- Pharmacological & Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| |
Collapse
|
47
|
Purewal JS, Doshi GM. RNAi in psoriasis: A melodic exploration of miRNA, shRNA, and amiRNA with a spotlight on siRNA. Eur J Pharmacol 2024; 985:177083. [PMID: 39481628 DOI: 10.1016/j.ejphar.2024.177083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/02/2024]
Abstract
Psoriasis (Pso) is an autoimmune inflammatory skin disease characterised by well-demarcated, red plaques covered in silver scales. It affects people of all ages and can be passed down through generations. Genetics play an important role in determining vulnerability to develop Pso. Several large-scale genome-wide association studies have identified over 80 genetic loci associated with Pso susceptibility. Gene expression can be regulated via RNA interference (RNAi). RNAi suppresses gene expression by degrading mRNA molecules. Since its discovery, RNAi has generated considerable excitement over its potential therapeutic benefits. RNAi is mediated by endogenous small RNA molecules like microRNA (miRNA) or exogenous small RNA molecules like small interfering RNA (siRNA), short hairpin RNA (shRNA), and artificial micro RNA (amiRNA). These small RNA molecules can silence a disease-related gene in a sequence-specific manner. Targeting RNAi pathways can help modify disease-related biological processes in various medical conditions, including autoimmune disorders. In Pso, RNAi can downregulate the expression of molecules involved in the pathophysiology of the disease. Significant progress has been made in the field of RNAi therapeutics. However, further research is needed to fine-tune the design and delivery of RNAi therapeutics in humans. In this review, we discuss various effectors of RNAi, some challenges related to RNAi therapeutics (emphasizing siRNA) and strategies to overcome these challenges. Furthermore, we have discussed some studies that employ RNAi therapeutics for Pso.
Collapse
|
48
|
Joveini S, Yarmohammadi F, Iranshahi M, Nikpoor AR, Askari VR, Attaranzadeh A, Etemad L, Taherzadeh Z. Distinct therapeutic effects of auraptene and umbelliprenin on TNF-α and IL-17 levels in a murine model of chronic inflammation. Heliyon 2024; 10:e40731. [PMID: 39687160 PMCID: PMC11648749 DOI: 10.1016/j.heliyon.2024.e40731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Objective To compare the anti-arthritic potential of orally administered auraptene (AUR) and umbellliprenin (UMB) in chronic inflammation by exploring the differential effect on regulating TNF-α and IL-17. Methods & materials Sixty male rats were divided into ten groups, and after confirming chronic inflammation, the treatment groups received AUR or UMB orally for 9 days. On day 16, histopathological changes were evaluated. Altered serum levels of the inflammatory cytokines TNF-α and IL-17 were examined as the underlying mechanisms. Results Administering AUR orally at 16 mM/kg caused a significant increase in body weight gain compared to the baseline (p < 0.05), while UMB at a dose of 64 mM/kg significantly reduced edema size (p < 0.01). TNF-α levels were significantly lower in all doses of AUR and UMB treatments compared to the arthritis control group (p < 0.05). Treatment with AUR at all relative doses resulted in a significant decrease in IL-17 levels compared to the arthritis control group (p < 0.05), whereas UMB treatment did not show a significant effect on IL-17 levels. Conclusion AUR and UMB regulate TNF-α and IL-17 differently; AUR inhibits both, showing broad therapeutic potential, while UMB specifically targets TNF-α, showing a specialized role.
Collapse
Affiliation(s)
- Saeid Joveini
- Department of Pharmacodynamy and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Yarmohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Reza Nikpoor
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Armin Attaranzadeh
- Department of Medical Genetics, Faculty of Medicines, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zhila Taherzadeh
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Targeted Drug Delivery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
49
|
Samuelsen A, Lehman E, Burrows P, Bonavia AS. Time-dependent variation in immunoparalysis biomarkers among patients with sepsis and critical illness. Front Immunol 2024; 15:1498974. [PMID: 39712015 PMCID: PMC11659229 DOI: 10.3389/fimmu.2024.1498974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction Immunoparalysis is a state of immune dysfunction characterized by a marked reduction in the immune system's responsiveness, often observed following severe infections, trauma, or critical illness. This study aimed to perform a longitudinal assessment of immune function over the initial two weeks following the onset of sepsis and critical illness. Methods We compared ex vivo-stimulated cytokine release from whole blood of critically ill patients to traditional markers of immunoparalysis, including monocyte Human Leukocyte Antigen (mHLA)-DR expression and absolute lymphocyte count (ALC). A total of 64 critically ill patients were recruited in a tertiary care academic medical setting, including 31 septic and 33 non-septic patients. Results While mHLA-DR expression significantly increased over time, this was primarily driven by the non-septic subset of critically ill patients. ALC recovery was more pronounced in septic patients. Ex vivo stimulation of blood from septic patients revealed significant increases in TNF and IL-6 production over time. However, interferon-gamma production varied depending on the ex vivo stimulant used, and after normalization of cytokine concentrations to lymphocyte counts, it did not show significant recovery over time from illness onset. No significant correlation was found between mHLA-DR expression and other immunoparalysis biomarkers. Discussion These findings suggest the need for more nuanced immune monitoring approaches beyond the traditional 'sepsis' versus 'non-sepsis' classifications in critically ill patients. Additionally, they provide further evidence of a potential window for targeted immunotherapy in the first weeks of critical illness.
Collapse
Affiliation(s)
- Abigail Samuelsen
- Department of Anesthesiology and Perioperative Medicine, Penn State Medical Center, Hershey, PA, United States
| | - Erik Lehman
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Parker Burrows
- Department of Anesthesiology and Perioperative Medicine, Penn State Medical Center, Hershey, PA, United States
| | - Anthony S Bonavia
- Department of Anesthesiology and Perioperative Medicine, Penn State Medical Center, Hershey, PA, United States
- Critical Illness and Sepsis Research Center, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
50
|
Xue B, Qualls C, Lanthiez A, Lu QY, Yang J, Lee RP, Neis P, Mao JT. The Effects of a Grape Seed Procyanidin Extract on Cytochrome P450 3A4 Activity and Inflammatory Mediators in the Lungs of Heavy Active and Former Smokers. Int J Mol Sci 2024; 25:13105. [PMID: 39684816 DOI: 10.3390/ijms252313105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Grape seed procyanidin extract (GSE) is widely used to promote cardiovascular health and has purported anti-inflammatory properties. Chronic inflammation in the lungs caused by environmental toxins such as tobacco smoking plays a pivotal role in lung cancer development. In a modified phase I lung cancer chemoprevention study conducted in heavy active and former smokers using leucoselect phytosome (LP), a standardized grape seed procyanidin extract complexed with soy phospholipids to enhance bioavailability, three months of LP treatment favorably modulated a variety of surrogate endpoint biomarkers, including markers of cell proliferation. In this correlative study, we further analyzed the effects of LP on cytochrome P450 3A4 (CYP3A4) activities by comparing the endogenous conversions of cortisol and cortisone to 6-beta-hydroxycortisol and 6-beta-hydroxycortisone, respectively, before and after LP treatment and the anti-inflammatory effects of LP in the lung microenvironment of these participants by comparing a profile of inflammatory cytokines and chemokines in matched pre- and post-treatment bronchoalveolar lavage (BAL) fluids. LP treatment did not significantly alter CYP3A4 activity, and three months of LP treatment significantly decreased tumor necrosis factor (TNF), C-C Motif Chemokine Ligand 3 (CCL3) and granzyme B in BAL fluids. Furthermore, post-LP-treatment BAL fluids significantly reduced migration/invasion of various human lung neoplastic cells in vitro. Our findings support the anti-inflammatory effects of GSE/LP in the lung microenvironment and its potential utility for reducing cancerizing forces, as well as driving forces for other common respiratory diseases such as chronic obstructive pulmonary disease and asthma, in the lungs of heavy former and active smokers.
Collapse
Affiliation(s)
- Bingye Xue
- Pulmonary, Critical Care and Sleep Section, New Mexico Veterans Administration Health Care System, University of New Mexico, Albuquerque, NM 87108, USA
| | - Clifford Qualls
- Biostatistics, Biomedical Research Institute of New Mexico, New Mexico Veterans Administration Health Care System, University of New Mexico, Albuquerque, NM 87108, USA
| | - Alexander Lanthiez
- Pulmonary and Critical Care Section, Veterans Administration San Diego Healthcare System, University of California San Diego, La Jolla, CA 92161, USA
| | - Qing-Yi Lu
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jieping Yang
- UCLA Center for Human Nutrition, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Ru-Po Lee
- UCLA Center for Human Nutrition, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Patricia Neis
- Pulmonary, Critical Care and Sleep Section, New Mexico Veterans Administration Health Care System, University of New Mexico, Albuquerque, NM 87108, USA
| | - Jenny T Mao
- Pulmonary, Critical Care and Sleep Section, New Mexico Veterans Administration Health Care System, University of New Mexico, Albuquerque, NM 87108, USA
- Pulmonary and Critical Care Section, Veterans Administration San Diego Healthcare System, University of California San Diego, La Jolla, CA 92161, USA
| |
Collapse
|