101
|
Bozó R, Flink LB, Ambrus B, Ghaffarinia A, Koncz B, Kui R, Gyulai R, Kemény L, Bata-Csörgő Z. The Expression of Cytokines and Chemokines Potentially Distinguishes Mild and Severe Psoriatic Non-Lesional and Resolved Skin from Healthy Skin and Indicates Different Stages of Inflammation. Int J Mol Sci 2024; 25:11292. [PMID: 39457071 PMCID: PMC11509107 DOI: 10.3390/ijms252011292] [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: 09/22/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
In the psoriatic non-lesional (PS-NL) skin, the tissue environment potentially influences the development and recurrence of lesions. Therefore, we aimed to investigate mechanisms involved in regulating tissue organization in PS-NL skin. Cytokine, chemokine, protease, and protease inhibitor levels were compared between PS-NL skin of patients with mild and severe symptoms and healthy skin. By comparing mild and severe PS-NL vs. healthy skin, differentially expressed cytokines and chemokines suggested alterations in hemostasis-related processes, while protease inhibitors showed no psoriasis severity-related changes. Comparing severe and mild PS-NL skin revealed disease severity-related changes in the expression of proteases, cytokines, and chemokines primarily involving methyl-CpG binding protein 2 (MECP2) and extracellular matrix organization-related mechanisms. Cytokine and chemokine expression in clinically resolved versus healthy skin showed slight interleukin activity, differing from patterns in mild and severe PS-NL skin. Immunofluorescence analysis revealed the severity-dependent nuclear expression pattern of MECP2 and decreased expression of 5-methylcytosine and 5-hydroxymethylcytosine in the PS-NL vs. healthy skin, and in resolved vs. healthy skin. Our results suggest distinct cytokine-chemokine signaling between the resolved and PS-NL skin of untreated patients with varying severities. These results highlight an altered inflammatory response, epigenetic regulation, and tissue organization in different types of PS-NL skin with possibly distinct, severity-dependent para-inflammatory states.
Collapse
Affiliation(s)
- Renáta Bozó
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (L.B.F.); (Z.B.-C.)
- HCEMM-USZ Skin Research Group, University of Szeged, H-6720 Szeged, Hungary
| | - Lili Borbála Flink
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (L.B.F.); (Z.B.-C.)
- HCEMM-USZ Skin Research Group, University of Szeged, H-6720 Szeged, Hungary
| | - Barbara Ambrus
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (L.B.F.); (Z.B.-C.)
| | - Ameneh Ghaffarinia
- HCEMM-USZ Skin Research Group, University of Szeged, H-6720 Szeged, Hungary
| | - Balázs Koncz
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary
- HCEMM-BRC Systems Immunology Research Group, H-6726 Szeged, Hungary
| | - Róbert Kui
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (L.B.F.); (Z.B.-C.)
| | - Rolland Gyulai
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (L.B.F.); (Z.B.-C.)
| | - Lajos Kemény
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (L.B.F.); (Z.B.-C.)
- HCEMM-USZ Skin Research Group, University of Szeged, H-6720 Szeged, Hungary
- HUN-REN-SZTE Dermatological Research Group, Hungarian Research Network, H-6720 Szeged, Hungary
| | - Zsuzsanna Bata-Csörgő
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (L.B.F.); (Z.B.-C.)
- HCEMM-USZ Skin Research Group, University of Szeged, H-6720 Szeged, Hungary
- HUN-REN-SZTE Dermatological Research Group, Hungarian Research Network, H-6720 Szeged, Hungary
| |
Collapse
|
102
|
Adejumo IO. Hypothetical proteins of chicken-isolated Limosilactobacillus reuteri subjected to in silico analyses induce IL-2 and IL-10. GENES & NUTRITION 2024; 19:21. [PMID: 39425027 PMCID: PMC11490116 DOI: 10.1186/s12263-024-00755-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/09/2024] [Indexed: 10/21/2024]
Abstract
Lactic acid bacteria (LAB) probiotics are health-promoting but their characteristics, safety profile and functional mechanisms are not fully understood. Hence, this study aimed to characterize some hypothetical proteins of the chicken-isolated Limosilactobacillus reuteri genome and unravel their IL-2 and IL-10-inducing potential to understand mechanisms of their immunological functionality for sustainable applications. The selected proteins were subjected to in silico analyses for transmembrane topology, sub-cellular localization, IL-2 and IL-10-inducing ability and IL-2 and IL-10 gene expression across various conditions. IL-2 and IL-10-inducing mutants were statistically analyzed using a one-way analysis of variance of a general linear model of SAS and statistical significance was set at p < 0.05. The analyzed proteins are stable under a wide temperature range. All the hypothetical proteins are IL-2 and IL-10-inducing but QHPv.2.12, QHPv.2.13 and QHPv.2.15 are non-immunogenic. The evaluated mutants are IL-2 and IL-10-inducers but QHPv.2.13 and QHPv.2.15 are not immunogenic. This study sheds light on understanding the functional mechanisms of chicken-isolated L. reuteri and suggests it or its proteins as potent candidates for feed additive and therapeutic purposes.
Collapse
|
103
|
Ji M, Ran X, Zuo H, Zhang Q. Novel Insights into the Kallikrein-Kinin System in Fulminant Myocarditis: Physiological Basis and Potential Therapeutic Advances. J Inflamm Res 2024; 17:7347-7360. [PMID: 39429854 PMCID: PMC11490248 DOI: 10.2147/jir.s488237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024] Open
Abstract
Fulminant myocarditis (FM) is characterized by rapid cardiac deterioration often instigated by an inflammatory cytokine storm. The kallikrein-kinin system (KKS) is a metabolic cascade known for releasing vasoactive kinins, such as bradykinin-related peptides, possessing diverse pharmacological activities that include inflammation, regulation of vascular permeability, endothelial barrier dysfunction, and blood pressure modulation. The type 1 and type 2 bradykinin receptors (B1R and B2R), integral components of the KKS system, mediate the primary biological effects of kinin peptides. This review aims to offer a comprehensive overview of the primary mechanisms of the KKS in FM, including an examination of the structural components, regulatory activation, and downstream signaling pathways of the KKS. Furthermore, it explores the involvement of the tissue kallikrein/B1R/inducible nitric oxide synthase (TK/B1R/iNOS) pathway in myocyte dysfunction, modulation of the immune response, and preservation of endothelial barrier integrity. The potential therapeutic advances targeting the inhibition of the KKS in managing FM will be discussed, providing valuable insights for the development of clinical treatment strategies.
Collapse
Affiliation(s)
- Mengmeng Ji
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Xiao Ran
- Department of Critical-Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Houjuan Zuo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Qin Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
104
|
Bufan B, Marčetić M, Djuretić J, Ćuruvija I, Blagojević V, Božić DD, Milutinović V, Janković R, Sopta J, Kotur-Stevuljević J, Arsenović-Ranin N. Evaluation of the Anti-Inflammatory/Immunomodulatory Effect of Teucrium montanum L. Extract in Collagen-Induced Arthritis in Rats. BIOLOGY 2024; 13:818. [PMID: 39452128 PMCID: PMC11505313 DOI: 10.3390/biology13100818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024]
Abstract
The anti-inflammatory/immunomodulatory effects of Teucrium montanum L. (TM), a plant distributed in the Mediterranean region, have been insufficiently examined. The effects of the TM ethanol extract were tested in a rat collagen-induced arthritis (CIA) model of rheumatoid arthritis. LC-MS was used for the phytochemical analysis of the TM extract. Dark Agouti rats were immunized with bovine type II collagen (CII) in incomplete Freund's adjuvant for CIA, and treated with 100 or 200 mg/kg of TM extract daily via oral administration. Clinical and histopathological evaluations and a flow cytometric analysis of the phenotypic and functional characteristics of splenocytes and draining lymph node cells were performed. The cytokines in the paw tissue culture supernatants and anti-CII antibodies in serum were determined by ELISA. The TM extract, with the dominant components verbascoside and luteolin 7-O-rutinoside, reduced the arthritic score and ankle joint inflammation in CIA rats, promoted the antioxidant profile in serum, and lowered pro-inflammatory TNF-α, IL-6 and IL-1β production. It suppressed the activation status of CD11b+ cells by lowering CD86, MHCII and TLR-4 expression, and promoted the Th17/T regulatory cell (Tregs) balance towards Tregs. A lower frequency of B cells was accompanied by a lower level of anti-CII antibodies in treated rats. These findings imply the favorable effect of TM extract on the clinical presentation of CIA, suggesting its anti-inflammatory/immunomodulatory action and potential therapeutic effect.
Collapse
Affiliation(s)
- Biljana Bufan
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia; (B.B.); (D.D.B.)
| | - Mirjana Marčetić
- Department of Pharmacognosy, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia; (M.M.); (V.M.)
| | - Jasmina Djuretić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia;
| | - Ivana Ćuruvija
- Institute of Virology, Vaccines and Sera “Torlak”, 11221 Belgrade, Serbia; (I.Ć.); (V.B.)
| | - Veljko Blagojević
- Institute of Virology, Vaccines and Sera “Torlak”, 11221 Belgrade, Serbia; (I.Ć.); (V.B.)
| | - Dragana D. Božić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia; (B.B.); (D.D.B.)
| | - Violeta Milutinović
- Department of Pharmacognosy, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia; (M.M.); (V.M.)
| | - Radmila Janković
- Institute of Pathology “Prof. dr Đorđe Joannović”, University of Belgrade-Faculty of Medicine, 11000 Belgrade, Serbia; (R.J.); (J.S.)
| | - Jelena Sopta
- Institute of Pathology “Prof. dr Đorđe Joannović”, University of Belgrade-Faculty of Medicine, 11000 Belgrade, Serbia; (R.J.); (J.S.)
| | - Jelena Kotur-Stevuljević
- Department of Medical Biochemistry, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia;
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia; (B.B.); (D.D.B.)
| |
Collapse
|
105
|
David-Vieira C, Carpinter BA, Bezerra-Bellei J, Machado L, Raimundo FO, Rodolphi CM, Renhe DC, Guedes IR, Gonçalves FMM, Pereira LPC, Ferreira MV, Nascimento HLDS, Neto AF, Gomes FR, Rocha VN, Castro JMA, Scopel KKG. Lung Damage Induced by Plasmodium berghei ANKA in Murine Model of Malarial Infection is Mitigated by Dietary Supplementation with DHA-Rich Omega-3. ACS Infect Dis 2024; 10:3607-3617. [PMID: 39303151 PMCID: PMC11474944 DOI: 10.1021/acsinfecdis.4c00482] [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: 06/12/2024] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are severe complications that can occur in infections caused by any Plasmodium species. Due to the high lethality rate and the lack of specific treatment for ALI/ARDS, studies aimed at understanding and searching for treatment strategies for such complications have been fundamental. Here, we investigated the protective role of dietary supplementation with DHA-rich fish oil against lung damage induced by Plasmodium berghei ANKA in a murine model. Our results demonstrated that alveolar vascular damage, lung edema, and histopathological alterations were significantly reduced in mice that received dietary supplementation compared to those that did not receive the supplementation. Furthermore, a significant reduction in the number of CD8+ T lymphocytes, in addition to reduced infiltration of inflammatory cells in the bronchoalveolar lavage fluid was also observed. High levels of IL-10, but not of TNF-α and IFN-γ, were also observed in infected mice that received the supplementation, along with a reduction in local oxidative stress. Together, the data suggest that dietary supplementation with DHA-rich fish oil in malarial endemic areas may help reduce lung damage resulting from the infection, thus preventing worsening of the condition.
Collapse
Affiliation(s)
- Carolina David-Vieira
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Barbara Albuquerque Carpinter
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Jéssica
Correia Bezerra-Bellei
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Letícia
Ferreira Machado
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Felipe Oliveira Raimundo
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Cinthia Magalhães Rodolphi
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Daniela Chaves Renhe
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Isabella Rodrigues
Nogueira Guedes
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Fernanda Mikaela Moreira Gonçalves
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Ludmila Ponce
Monken Custódio Pereira
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | | | - Haroldo Lobo dos Santos Nascimento
- Research
Centre of Pathology and Veterinary Histology, Department of Veterinary
Medicine, Federal University of Juiz de
Fora, Juiz de
Fora 36036-900, Brazil
| | - Adolfo Firmino Neto
- Research
Centre of Pathology and Veterinary Histology, Department of Veterinary
Medicine, Federal University of Juiz de
Fora, Juiz de
Fora 36036-900, Brazil
| | | | - Vinicius Novaes Rocha
- Research
Centre of Pathology and Veterinary Histology, Department of Veterinary
Medicine, Federal University of Juiz de
Fora, Juiz de
Fora 36036-900, Brazil
| | - Juciane Maria
de Andrade Castro
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Kézia Katiani Gorza Scopel
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| |
Collapse
|
106
|
Cao H, Shi C, Aihemaiti Z, Dai X, Wang F, Wang S. Association between circulating inflammatory proteins and benign prostatic disease: a Mendelian randomization study. Sci Rep 2024; 14:23667. [PMID: 39390078 PMCID: PMC11467427 DOI: 10.1038/s41598-024-74737-2] [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: 06/21/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
Previous research has suggested that circulating inflammatory proteins are associated with benign prostatic disease (BPD). This Mendelian randomization (MR) study was conducted to further investigate the causal relationship between 91 inflammatory proteins and BPD. Genome-wide association study (GWAS) summarized data of benign prostatic hyperplasia (BPH) and prostatitis were obtained from the FinnGen Biobank. The latest study offered the GWAS data on 91 proteins related to inflammation. We performed a bidirectional MR to investigate the causal association between inflammatory proteins and BPD. The outcomes of the IVW method indicated that decreased levels of circulating interleukin-17 C (IL-17 C) (OR = 0.92, 95%CI = 0.85-0.99, p-value = 0.0344) were suggestively associated with a higher risk of BPH and elevated levels of interleukin-10 receptor subunit alpha (IL-10RA) (OR = 1.24, 95%CI = 1.05-1.47, p-value = 0.0132) and urokinase-type plasminogen activator (uPA) (OR = 1.13, 95%CI = 1.00-1.28, p-value = 0.0421) were suggestively related to a higher risk of prostatitis. Furthermore, reverse MR revealed that BPH may promote the expression of circulating factors, including natural killer cell receptor 2B4 (CD244) (OR = 1.07, 95%CI = 1.01-1.13, p-value = 0.0192), T-cell surface glycoprotein CD6 isoform (CD6) (OR = 1.07, 95%CI = 1.01-1.13, p-value = 0.0192), and leukemia inhibitory factor receptor (LIF-R) (OR = 1.07, 95%CI = 1.01-1.15, p-value = 0.0163). Moreover, the results of sensitivity analyses indicate that heterogeneity and horizontal pleiotropy are unlikely to distort the findings. The results of this study indicate a potential association between circulating inflammatory proteins and BPD, which may become new diagnostic indicators or drug targets for clinical application in the prevention and treatment of BPD. However, further investigation is required.
Collapse
Affiliation(s)
- Hongliang Cao
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Chengdong Shi
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zulipikaer Aihemaiti
- Department of Urology, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830002, China
| | - Xianyu Dai
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Fulin Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Song Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
107
|
Gu Q, Draheim M, Planchais C, He Z, Mu F, Gong S, Shen C, Zhu H, Zhivaki D, Shahin K, Collard JM, Su M, Zhang X, Mouquet H, Lo-Man R. Intestinal newborn regulatory B cell antibodies modulate microbiota communities. Cell Host Microbe 2024; 32:1787-1804.e9. [PMID: 39243760 DOI: 10.1016/j.chom.2024.08.010] [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: 02/16/2024] [Revised: 07/08/2024] [Accepted: 08/12/2024] [Indexed: 09/09/2024]
Abstract
The role of immunoglobulins produced by IL-10-producing regulatory B cells remains unknown. We found that a particular newborn regulatory B cell population (nBreg) negatively regulates the production of immunoglobulin M (IgM) via IL-10 in an autocrine manner, limiting the intensity of the polyreactive antibody response following innate activation. Based on nBreg scRNA-seq signature, we identify these cells and their repertoire in fetal and neonatal intestinal tissues. By characterizing 205 monoclonal antibodies cloned from intestinal nBreg, we show that newborn germline-encoded antibodies display reactivity against bacteria representing six different phyla of the early microbiota. nBreg-derived antibodies can influence the diversity and the cooperation between members of early microbial communities, at least in part by modulating energy metabolism. These results collectively suggest that nBreg populations help facilitate early-life microbiome establishment and shed light on the paradoxical activities of regulatory B cells in early life.
Collapse
Affiliation(s)
- Qisheng Gu
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Unit of Immunity and Pediatric Infectious Diseases, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China; Université Paris Cite, Paris, France
| | - Marion Draheim
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Unit of Immunity and Pediatric Infectious Diseases, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Cyril Planchais
- Humoral Immunology Unit, Institut Pasteur, Université Paris Cite, INSERM U1222, Paris, France
| | - Zihan He
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Unit of Immunity and Pediatric Infectious Diseases, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Fan Mu
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Unit of Immunity and Pediatric Infectious Diseases, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Shijie Gong
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Unit of Immunity and Pediatric Infectious Diseases, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Chun Shen
- Children's Hospital of Fudan University, Shanghai, China
| | - Haitao Zhu
- Children's Hospital of Fudan University (Xiamen Branch), Xiamen Children's Hospital, Xiamen, China
| | - Dania Zhivaki
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Khashayar Shahin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan Microbiome Center, and Human Phenome Institute, Fudan University, Shanghai, China
| | - Jean-Marc Collard
- Enteric Bacterial Pathogens Unit & French National Reference Center for Escherichia Coli, Shigella and Salmonella, Institut Pasteur, Paris, France
| | - Min Su
- Obstetrics department, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoming Zhang
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Unit of Innate Defense and Immune Modulation, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Hugo Mouquet
- Humoral Immunology Unit, Institut Pasteur, Université Paris Cite, INSERM U1222, Paris, France.
| | - Richard Lo-Man
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Unit of Immunity and Pediatric Infectious Diseases, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China; Université Paris Cite, Paris, France.
| |
Collapse
|
108
|
Arfath Y, Kotra T, Faizan MI, Akhtar A, Abdullah ST, Ahmad T, Ahmed Z, Rayees S. TRPV4 facilitates the reprogramming of inflamed macrophages by regulating IL-10 production via CREB. Inflamm Res 2024; 73:1687-1697. [PMID: 39101955 DOI: 10.1007/s00011-024-01923-3] [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: 03/05/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Transient receptor potential vanilloid type 4 (TRPV4) is a versatile ion channel with diverse roles in immune cells, including macrophages. While its function in inflammation remains debated, we investigated its role in regulating IL-10 production and its impact on macrophage reprogramming during inflammation. METHODS We investigated the connection between TRPV4 activation and CREB-mediated IL-10 production during inflammation. Notably, this signaling pathway was found to reprogram macrophages and enhance their ability to resist inflammatory damage. The experiments were conducted on primary macrophages and were further corroborated by animal studies. RESULTS In response to TRPV4 activation during inflammation, we observed a significant increase in intracellular Ca2+ levels, which triggered the activation of the transcription factor CREB, subsequently upregulating IL-10 production. This IL-10 played a pivotal role in reprogramming macrophages to withstand inflammatory damage. Using a mouse model of acute lung injury (ALI), we confirmed that TRPV4 activation during ALI led to IL-10 secretion, but this increase did not significantly contribute to inflammation resolution. Moreover, we found that TRPV4 prevented the accumulation of dysfunctional mitochondria in macrophages through the CREB-IL-10 axis during inflammation. Suppression of CREB or TRPV4 inhibition exacerbated mitochondrial dysfunction, while treatment with recombinant IL-10 mitigated these effects. Additionally, IL-10 induced mitophagy and cleared dysfunctional mitochondria in LPS-exposed cells. CONCLUSION Our study highlights the essential role of TRPV4 in regulating IL-10 production and mitochondrial health in macrophages during inflammation. These findings contribute to understand the role of TRPV4 in immune responses and suggest potential therapeutic targets for modulating inflammation-induced cellular dysfunction.
Collapse
Affiliation(s)
- Yassir Arfath
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Tusharika Kotra
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Md Imam Faizan
- Multidisciplinary Centre for Advanced Research and Studies, JMI, New Delhi, 110025, India
| | - Areej Akhtar
- Multidisciplinary Centre for Advanced Research and Studies, JMI, New Delhi, 110025, India
| | - Sheikh Tasduq Abdullah
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advanced Research and Studies, JMI, New Delhi, 110025, India
| | - Zabeer Ahmed
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.
| | - Sheikh Rayees
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu, 180001, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
109
|
Yasavoli‐Sharahi H, Shahbazi R, Alsadi N, Robichaud S, Kambli D, Izadpanah A, Mohsenifar Z, Matar C. Edodes Cultured Extract Regulates Immune Stress During Puberty and Modulates MicroRNAs Involved in Mammary Gland Development and Breast Cancer Suppression. Cancer Med 2024; 13:e70277. [PMID: 39382253 PMCID: PMC11462599 DOI: 10.1002/cam4.70277] [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: 07/16/2024] [Revised: 09/09/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Immune stressors, such as lipopolysaccharides (LPS), profoundly affect microbiota balance, leading to gut dysbiosis. This imbalance disrupts the metabolic phenotype and structural integrity of the gut, increasing intestinal permeability. During puberty, a critical surge in estrogen levels is crucial for mammary gland development. However, inflammation originating from the gut in this period may interfere with this development, potentially heightening breast cancer risk later. The long-term effects of pubertal inflammation on mammary development and breast cancer risk are underexplored. Such episodes can dysregulate cytokine levels and microRNA expression, altering mammary cell gene expression, and predisposing them to tumorigenesis. METHODS This study hypothesizes that prebiotics, specifically Lentinula edodes Cultured Extract (AHCC), can counteract LPS's adverse effects. Using BALB/c mice, an acute LPS dose was administered at puberty, and breast cancer predisposition was assessed at 13 weeks. Cytokine and tumor-related microRNA levels, tumor development, and cancer stem cells were explored through immunoassays and qRT-PCR. RESULTS Results show that LPS induces lasting effects on cytokine and microRNA expression in mammary glands and tumors. AHCC modulates cytokine expression, including IL-1β, IL-17A/F, and IL-23, and mitigates LPS-induced IL-6 in mammary glands. It also regulates microRNA expression linked to tumor progression and suppression, particularly counteracting the upregulation of oncogenic miR-21, miR-92, and miR-155. Although AHCC slightly alters some tumor-suppressive microRNAs, these changes are modest, highlighting a complex regulatory role that warrants further study. CONCLUSION These findings underscore the potential of dietary interventions like AHCC to mitigate pubertal LPS-induced inflammation on mammary gland development and tumor formation, suggesting a preventive strategy against breast cancer.
Collapse
Affiliation(s)
- Hamed Yasavoli‐Sharahi
- Cellular and Molecular Medicine Department, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Roghayeh Shahbazi
- Cellular and Molecular Medicine Department, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Nawal Alsadi
- Cellular and Molecular Medicine Department, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Samuel Robichaud
- Department of PathologyUniversity of MontrealMontrealQuebecCanada
| | - Darshan Babu Kambli
- Cellular and Molecular Medicine Department, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Amirhossein Izadpanah
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Zhaleh Mohsenifar
- Department of PathologySchool of Medicine, Shahid Beheshti University of Medical SciencesTehranIran
| | - Chantal Matar
- Cellular and Molecular Medicine Department, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- School of Nutrition Sciences, Faculty of Health SciencesUniversity of OttawaOttawaOntarioCanada
| |
Collapse
|
110
|
Bakhashab S, Banafea GH, Ahmed F, Alsolami R, Schulten HJ, Gauthaman K, Naseer MI, Pushparaj PN. Acute and prolonged effects of interleukin-33 on cytokines in human cord blood-derived mast cells. Immunol Lett 2024; 269:106908. [PMID: 39151731 DOI: 10.1016/j.imlet.2024.106908] [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: 05/02/2024] [Revised: 07/11/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
Mast cells are multifaceted cells localized in tissues and possess various surface receptors that allow them to respond to inner and external threat signals. Interleukin-33 (IL-33) is a cytokine released by structural cells in response to parasitic infections, mechanical damage, and cell death. IL-33 can activate mast cells, causing them to release an array of mediators. This study aimed to identify the different cytokines released by human cord blood-derived mast cells (hCBMCs) in response to acute and prolonged stimulation with IL-33. For this purpose, a hCBMC model was established and stimulated with 10 ng and 20 ng of recombinant human IL-33 (rhIL-33) for 6 h and 24 h. Total RNA was hybridized using a high-density oligonucleotide microarray. A multiplex assay was performed to assess the released cytokines. Acute exposure to rhIL-33 increased the expression of IL-1α, IL-1β, IL-6, and IL-13, whereas prolonged exposure increased the expression of IL-5 and IL-10, and cytokines were detected in the culture supernatant. WebGestalt analysis revealed that rhIL-33 induces pathways and biological processes related to the immune system and the acute inflammatory response. This study demonstrates that rhIL-33 can activate hCBMCs to release pro- and anti-inflammatory cytokines, eliciting distinct acute and prolonged responses unique to hCBMCs.
Collapse
Affiliation(s)
- Sherin Bakhashab
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia; Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Ghalya H Banafea
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Farid Ahmed
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Reem Alsolami
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hans-Juergen Schulten
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Kalamegam Gauthaman
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Muhammad Imran Naseer
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia; Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
111
|
Song Y, Sun X, Shen L, Qu Z, Yin J, Wang Z, Zhang H. Genes of cancer-related fatigue: a scoping review. Front Oncol 2024; 14:1446321. [PMID: 39372868 PMCID: PMC11449716 DOI: 10.3389/fonc.2024.1446321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 08/08/2024] [Indexed: 10/08/2024] Open
Abstract
Background Cancer-related fatigue (CRF) is a prevalent adverse effect experienced by cancer patients while receiving and after treatment, impacting as many as 90% of individuals. Although CRF is common, the genetic processes responsible for it and their influence on individual vulnerability are not well understood and are still being investigated. Objective The primary objective of this scoping review is to identify and assess genes linked to the vulnerability and severity of CRF. This will help us better understand the genetic factors involved and assist in developing targeted nursing treatments in clinical settings. Methods This review followed the PRISMA guidelines. A comprehensive search was performed in databases, such as PubMed, EMBASE, Web of Science, Cochrane Library, SinoMed, CNKI, and VIP, encompassing genetic association studies on CRF published up to February 25, 2024. The JBI Critical Appraisal Tools were used to assess the quality of observational studies. Results This evaluation encompassed a comprehensive analysis of 14 studies that involved 3,254 patients. The results indicate strong connections between CRF and various inflammatory cytokines (IL-4, IL-6, IL-8, IL-10, IL-1β), tumor necrosis factor-alpha (TNF-α), catechol-O-methyltransferase (COMT), and circadian rhythm genes (CLOCK, PER). Conclusion This scoping review emphasizes the significant genetic factor in CRF, with multiple genes showing distinct effects on cancer fatigue symptoms. Identifying these genes enhances our comprehension of CRF and unveils novel avenues for cancer treatment approaches. Future research should prioritize conducting cohort studies to monitor alterations in gene expression pre- and post-treatment, hence improving individualized medicinal strategies in oncology.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hongshi Zhang
- College of Nursing, Changchun University of Chinese Medicine,
Changchun, Jilin, China
| |
Collapse
|
112
|
Shaikh A, Gangaplara A, Kone A, Almengo K, Kabore MD, Ali MA, Xu X, Saxena A, Lopez-Ocasio M, McCoy JP, Fitzhugh CD. Galectin-1 is associated with hematopoietic cell engraftment in murine MHC-mismatched allotransplantation. Front Immunol 2024; 15:1411392. [PMID: 39351218 PMCID: PMC11439684 DOI: 10.3389/fimmu.2024.1411392] [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: 04/02/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Haploidentical hematopoietic cell transplantation (haplo-HCT) is associated with an increased risk of allograft rejection. Here, we employed a major histocompatibility complex (MHC)-mismatched allogeneic HCT (allo-HCT) murine model to better understand the role of Gal-1 in immune tolerance. Transplanted mice were classified into either rejected or engrafted based on donor chimerism levels. We noted significantly higher frequencies of CD4+ T cells, CD8+ T cells, natural killer cells, IFN-γ and TNF-α producing CD4+ T cells, and IFN-γ producing dendritic cells and macrophages in rejected mice. Conversely, we found significantly increased frequencies of regulatory T cells (Tregs), predominantly Helios+, IL-10-producing CD4+ T cells, type 1 regulatory (Tr1) cells, and the proportion of Tr1+Gal-1+ cells in engrafted mice. Further, Gal-1 specific blockade in Tregs reduced suppression of effector T cells in engrafted mice. Lastly, effector T cells from engrafted mice were more prone to undergo apoptosis. Collectively, we have shown that Gal-1 may favor HSC engraftment in an MHC-mismatched murine model. Our results demonstrate that Gal-1-expressing Tregs, especially at earlier time points post-transplant, are associated with inducing immune tolerance and stable mixed chimerism after HCT.
Collapse
Affiliation(s)
- Ahmad Shaikh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- Department of Biology, The Catholic University of America, Washington, DC, United States
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Arunakumar Gangaplara
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- Miltenyi Biotec, Research and Development, Gaithersburg, MD, United States
| | - Abdoul Kone
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Katherine Almengo
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mariama D. Kabore
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mohamed A.E. Ali
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xin Xu
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ankit Saxena
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Maria Lopez-Ocasio
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - J. Philip McCoy
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Courtney D. Fitzhugh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
113
|
Das A, Wang X, Devonshire K, Lyons EJ, Popescu I, Zhou Z, Li J, Sembrat J, Pilewski J, Zou C, Alder JK, Chen BB, Snyder ME, McDyer JF. IL-10 Is Critical for Regulation of Cytotoxic CD4+NKG7+ T Cells in Lung Allograft Rejection but Is Not Required for Allograft Acceptance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:898-905. [PMID: 39072690 DOI: 10.4049/jimmunol.2400279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024]
Abstract
Lung transplant remains the primary therapeutic option for patients with end-stage lung disease, but long-term survival rates remain suboptimal compared with other solid organ transplants. Acute cellular rejection (ACR) is a significant challenge in lung transplant recipients, with T cell-mediated mechanisms playing a major role. IL-10 is known for its immunoregulatory function, although its specific role in lung allograft rejection remains unclear. Using the mouse orthotopic lung transplant model, we investigated the role of IL-10 in regulating alloeffector T cell responses. Unexpectedly, we found that IL-10 was not required for early costimulation blockade-induced allograft acceptance. However, IL-10 deficiency or blockade resulted in increased CD4+ T cell numbers, proliferation, graft infiltration, and alloeffector responses. In the absence of IL-10, CD4+ T cell responses predominated over CD8 responses during ACR in contrast to wild-type mice. Type 1 immunity (IFN-γ) responses along with elevated CD4+NKG7+ and CD4+CD107a+ responses predominated during ACR, highlighting a critical regulatory role for IL-10 in modulating CD4+ T cell alloimmune responses. We further demonstrated increased colocalization of NKG7 and CD107a in CD4+ T cells from IL-10-deficient allografts, suggesting coordination in cytotoxic activity. Together, our findings highlight a critical role for IL-10 in regulation of cytotoxic CD4+NKG7+ T cells, an effector population that needs further investigation to elucidate their role in lung allograft rejection.
Collapse
Affiliation(s)
- Antu Das
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Xingan Wang
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Kaitlyn Devonshire
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Emily J Lyons
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Iulia Popescu
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Zihe Zhou
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jingmei Li
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - John Sembrat
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Joseph Pilewski
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Chunbin Zou
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jonathan K Alder
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Bill B Chen
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Aging Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Mark E Snyder
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - John F McDyer
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
114
|
Fernandes Q, Ansari AW, Makni-Maalej K, Merhi M, Dermime S, Ahmad A, Uddin S. Interleukin 10: Bridging the chasms in the immune landscape of multiple myeloma. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 391:199-222. [PMID: 39939076 DOI: 10.1016/bs.ircmb.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Abstract
Multiple myeloma (MM) is a complex hematologic malignancy characterized by the abnormal proliferation of plasma cells in the bone marrow, leading to significant clinical challenges and a high burden of morbidity and mortality. Interleukin 10 (IL-10), a cytokine with potent anti-inflammatory properties, has emerged as a critical player in the pathobiology of MM. This work delves into the multifaceted role of IL-10 in MM, exploring its contributions to tumor growth, immune evasion, and drug resistance. Here, we examine IL-10's interactions with various immune cells within the bone marrow microenvironment and its potential as a circulatory biomarker for MM. Furthermore, we particularly lay emphasis on the prognostic and diagnostic implications of IL-10 levels in MM patients and evaluate the therapeutic prospects of targeting IL-10 in MM treatment regimens. By synthesizing current research, this review aims to enhance the understanding of IL-10 as a circulatory biomarker in MM and to highlight novel avenues for therapeutic intervention, thereby translating to improved clinical outcomes for MM patient.
Collapse
Affiliation(s)
- Queenie Fernandes
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Abdul W Ansari
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Karama Makni-Maalej
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; College of Health Sciences, Qatar University, Doha, Qatar
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Department of Bioengineering, Integral University, Lucknow, Uttar Pradesh, India
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Laboratory of Animal Research Center, Qatar University, Doha, Qatar; Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
115
|
Zhou JQ, Liu ZX, Zhong HF, Liu GQ, Ding MC, Zhang Y, Yu B, Jiang N. Single nucleotide polymorphisms in the development of osteomyelitis and prosthetic joint infection: a narrative review. Front Immunol 2024; 15:1444469. [PMID: 39301021 PMCID: PMC11410582 DOI: 10.3389/fimmu.2024.1444469] [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: 06/05/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024] Open
Abstract
Currently, despite advancements in diagnostic and therapeutic modalities, osteomyelitis and prosthetic joint infection (PJI) continue to pose significant challenges for orthopaedic surgeons. These challenges are primarily attributed to the high degree of heterogeneity exhibited by these disorders, which are influenced by a combination of environmental and host factors. Recent research efforts have delved into the pathogenesis of osteomyelitis and PJI by investigating single nucleotide polymorphisms (SNPs). This review comprehensively summarizes the current evidence regarding the associations between SNPs and the predisposition to osteomyelitis and PJI across diverse populations. The findings suggest potential linkages between SNPs in genes such as IL-1, IL-6, IFN-γ, TNF-α, VDR, tPA, CTSG, COX-2, MMP1, SLC11A1, Bax, NOS2, and NLRP3 with the development of osteomyelitis. Furthermore, SNPs in genes like IL-1, IL-6, TNF-α, MBL, OPG, RANK, and GCSFR are implicated in susceptibility to PJI. However, it is noted that most of these studies are single-center reports, lacking in-depth mechanistic research. To gain a more profound understanding of the roles played by various SNPs in the development of osteomyelitis and PJI, future multi-center studies and fundamental investigations are deemed necessary.
Collapse
Affiliation(s)
- Jia-Qi Zhou
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Orthopaedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Functional Repair of Bone Defects and Biomaterials, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zi-Xian Liu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Hong-Fa Zhong
- Department of Trauma Emergency Center, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
| | - Guan-Qiao Liu
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ming-Cong Ding
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Yu Zhang
- Department of Orthopaedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Functional Repair of Bone Defects and Biomaterials, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bin Yu
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Nan Jiang
- Division of Orthopaedics & Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Trauma Emergency Center, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, China
| |
Collapse
|
116
|
Wang S, Xu Y, Wang L, Lin J, Xu C, Zhao X, Zhang H. TolDC Restores the Balance of Th17/Treg via Aryl Hydrocarbon Receptor to Attenuate Colitis. Inflamm Bowel Dis 2024; 30:1546-1555. [PMID: 38431309 DOI: 10.1093/ibd/izae022] [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: 07/27/2023] [Indexed: 03/05/2024]
Abstract
BACKGROUND Tolerogenic dendritic cells (TolDCs) have been evidenced to trigger regulatory T cell's (Treg's) differentiation and be involved in the pathogenesis of Crohn's disease (CD). Aryl hydrocarbon receptor (AhR) plays a crucial role in the differentiation of TolDCs, although the mechanism remains vague. This study aimed to evaluate the role of AhR in TolDCs formation, which may affect Th17/Treg balance in CD. METHODS Colon biopsy specimens were obtained from healthy controls and patients with CD. Wild type (WT) and AhR-/- mice were induced colitis by drinking dextran sulphate sodium (DSS) with or without 6-formylindolo 3,2-b carbazole (FICZ) treatment. Wild type and AhR-/- bone marrow-derived cells (BMDCs) were cultured under TolDCs polarization condition. Ratios of DCs surface markers were determined by flow cytometry. Enzyme-linked immunosorbent assay (ELISA) was performed to quantify the levels of interleukin (IL)-1β, transforming growth factor (TGF)-β and IL-10. Tolerogenic dendritic cells differentiated from BMDCs of WT or AhR-/- mice were adoptively transferred to DSS-induced WT colitis mice. RESULTS Patients with CD showed less AhR expression and activation in their inflamed colon regions. Compared with WT mice, AhR-/- mice experienced more severe colitis. Tolerogenic dendritic cells and Tregs were both decreased in the colon of AhR-/- colitis mice, while Th17 cells were upregulated. In vitro, compared with WT DCs, AhR-deficient DCs led to less TolDC formation. Furthermore, intestinal inflammation in WT colitis mice, which transferred with AhR-/- TolDCs, showed no obvious improvement compared with those transferred with WT TolDCs, as evidenced by no rescues of Th17/Treg balance. CONCLUSIONS Activation of AhR attenuates experimental colitis by modulating the balance of TolDCs and Th17/Treg. The AhR modulation of TolDCs may be a viable therapeutic approach for CD.
Collapse
Affiliation(s)
- Shu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Ying Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Lu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Junjie Lin
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Chenjing Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Xiaojing Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Hongjie Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| |
Collapse
|
117
|
Pereira CH, Martins AFL, Morais MO, de Sousa-Neto SS, da Silva ACG, Arantes DAC, De Oliveira Moreira VHL, Valadares MC, Freitas NMA, Leles CR, Mendonça EF. Oral mucositis management with photobiomodulation, Bidens pilosa L. (Asteraceae) and Curcuma longa L. (Zingiberaceae), the FITOPROT herbal medicine, and its influence on inflammatory cytokine levels: a randomized clinical trial. Support Care Cancer 2024; 32:628. [PMID: 39223301 DOI: 10.1007/s00520-024-08842-3] [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: 08/10/2023] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE This randomized clinical trial aimed to compare the effects of a mucoadhesive formula, containing curcuminoids from Curcuma longa L. and glycerinated extract of Bidens pilosa L. (FITOPROT), associated with photobiomodulation (PBM), and of PBM exclusively, on the incidence of oral mucositis (OM)-induced by radiotherapy (RT) in the head and neck region, and the salivary expression of inflammatory cytokines, in patients with head neck cancer. METHODS Patients were randomly assigned into two intervention groups-FITOPROT + PBM (n = 25) or PBM (n = 27). PBM protocol comprised a wavelength of 660 nm, 25 mW, 0.25 J/point, and daily irradiation from the first until the last day of RT. FITOPROT was gargled twice a day. All patients underwent a preventive oral care program throughout the study. OM degree, salivary concentration of nitrite, and inflammatory (IL-1, TNFα, IL-6, IL-8, and IL-12p70), and anti-inflammatory (IL-10) cytokines were assessed at baseline, and at the 7th, 14th, 21st, and 30th RT sessions. RESULTS There were no differences in the OM degree between groups, but the RT dose significantly affected the OM. The RT significantly affected the salivary nitrite, TNFα, IL-1β, and IL-10 concentrations. CONCLUSION FITOPROT associated with PBM showed limited effects on preventing the incidence of severe OM compared to PBM alone. However, FITOPROT + PBM may be associated with nitrite and cytokine balance, which may contribute to the occurrence of fewer cases of severe OM. TRIAL REGISTRATION Brazilian Clinical Trials database (ReBEC; RBR-9vddmr), registered UTN code: U1111-1193-2066, registered in August 8th, 2017.
Collapse
Affiliation(s)
- Carlos Henrique Pereira
- Araujo Jorge Cancer Hospital, R. 239, 206 - Setor Universitário, Goiânia, Goiás, CEP 74175-120, Brazil
| | - Allisson Filipe Lopes Martins
- Department of Oral Diagnosis, School of Dentistry, Universidade Evangélica de Goiás, Dentistry School, Anápolis, Goiás, CEP 75083-515, Brazil
| | - Marília Oliveira Morais
- Araujo Jorge Cancer Hospital, R. 239, 206 - Setor Universitário, Goiânia, Goiás, CEP 74175-120, Brazil
| | - Sebastião Silvério de Sousa-Neto
- Laboratory of Oral Pathology, School of Dentistry, Universidade Federal de Goiás, Avenida Universitária Esquina Com 1ª Avenida, S/N. Setor Universitário, Goiânia, Goiás, CEP 74605-220, Brazil
| | - Artur Christian Garcia da Silva
- Laboratory of Education and Research in In Vitro Toxicology, Tox In, Faculty of Pharmacy, Universidade Federal de Goiás, Rodovia R2, N. 3.061, Campus Samambaia, Goiânia, Goiás, CEP 74605.170, Brazil
| | - Diego Antonio Costa Arantes
- Laboratory of Oral Pathology, School of Dentistry, Universidade Federal de Goiás, Avenida Universitária Esquina Com 1ª Avenida, S/N. Setor Universitário, Goiânia, Goiás, CEP 74605-220, Brazil
| | - Victor Hugo Lopes De Oliveira Moreira
- Laboratory of Oral Pathology, School of Dentistry, Universidade Federal de Goiás, Avenida Universitária Esquina Com 1ª Avenida, S/N. Setor Universitário, Goiânia, Goiás, CEP 74605-220, Brazil
| | - Marize Campos Valadares
- Laboratory of Education and Research in In Vitro Toxicology, Tox In, Faculty of Pharmacy, Universidade Federal de Goiás, Rodovia R2, N. 3.061, Campus Samambaia, Goiânia, Goiás, CEP 74605.170, Brazil
| | - Nilceana Maya Aires Freitas
- Department of Radiotherapy, Araujo Jorge Cancer Hospital, R. 239, 206-Setor Universitário, Goiânia, Goiás, CEP 74175-120, Brazil
| | - Cláudio Rodrigues Leles
- Department of Prevention and Oral Rehabilitation, School of Dentistry, Universidade Federal de Goiás, Avenida Universitária Esquina Com 1ª Avenida, S/N. Setor Universitário, Goiânia, Goiás, CEP 74605-220, Brazil
| | - Elismauro Francisco Mendonça
- Laboratory of Oral Pathology, School of Dentistry, Universidade Federal de Goiás, Avenida Universitária Esquina Com 1ª Avenida, S/N. Setor Universitário, Goiânia, Goiás, CEP 74605-220, Brazil.
| |
Collapse
|
118
|
Nguyen T, Kuhn K, Bolt M, Duffy K, Bradford AP, Santoro N. Analysis of Inflammatory Markers in Response to Induction of Reprometabolic Syndrome by a Eucaloric High Fat Diet in Normal Weight Women. Reprod Sci 2024; 31:2820-2828. [PMID: 38710978 DOI: 10.1007/s43032-024-01586-9] [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: 01/18/2024] [Accepted: 04/29/2024] [Indexed: 05/08/2024]
Abstract
Obesity is associated with chronic low-level inflammation and is known to contribute to metabolic dysfunction and hypogonadotropic hypogonadism, which we have previously termed the 'Reprometabolic Syndrome.' To investigate potential factors involved in obesity-related reproductive endocrine dysfunction, we conducted a secondary analysis of inflammatory markers in a sample of normal weight women exposed to a one-month eucaloric high-fat diet (HFD), which, as reported earlier, induced the relative hypogonadotropic hypogonadism characteristic of Reprometabolic Syndrome. Eighteen healthy women with a BMI between 18.0-24.9 kg/m2 and regular menstrual cycles participated in the study. Frequent blood sampling was performed during the early follicular phase before and after the one-month eucaloric HFD intervention (48% of calories from fat). Serum samples pooled from each participant were analyzed using immunoassay to measure levels of 30 cytokines, interleukins, and chemokines. Differences between pre- and post-HFD intervention measures were examined by one-sample t-tests. Exposure to the eucaloric HFD did not result in changes in body weight. HFD-induction of Reprometabolic Syndrome in normal weight women was associated with a significant elevation only in the anti- inflammatory cytokine IL-10 (p = 0.04). Eotaxin, IL-6 and MIP-1β also increased in response to the HFD, but not statistically significantly (p = 0.07). Results suggest that the increase in multiple inflammatory markers, typically associated with obesity, are not primary mediators of the relative hypogonadotropic hypogonadism of Reprometabolic Syndrome. Clinical Trials Registration Number: NCT02653092; Date of Registration: January 6, 2016.
Collapse
Affiliation(s)
- Thy Nguyen
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Katherine Kuhn
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Matthew Bolt
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, 80045, USA
| | - Katelyn Duffy
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Andrew P Bradford
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| | - Nanette Santoro
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| |
Collapse
|
119
|
Mihalić A, Železnjak J, Lisnić B, Jonjić S, Juranić Lisnić V, Brizić I. Immune surveillance of cytomegalovirus in tissues. Cell Mol Immunol 2024; 21:959-981. [PMID: 39134803 PMCID: PMC11364667 DOI: 10.1038/s41423-024-01186-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/14/2024] [Indexed: 09/01/2024] Open
Abstract
Cytomegalovirus (CMV), a representative member of the Betaherpesvirinae subfamily of herpesviruses, is common in the human population, but immunocompetent individuals are generally asymptomatic when infected with this virus. However, in immunocompromised individuals and immunologically immature fetuses and newborns, CMV can cause a wide range of often long-lasting morbidities and even death. CMV is not only widespread throughout the population but it is also widespread in its hosts, infecting and establishing latency in nearly all tissues and organs. Thus, understanding the pathogenesis of and immune responses to this virus is a prerequisite for developing effective prevention and treatment strategies. Multiple arms of the immune system are engaged to contain the infection, and general concepts of immune control of CMV are now reasonably well understood. Nonetheless, in recent years, tissue-specific immune responses have emerged as an essential factor for resolving CMV infection. As tissues differ in biology and function, so do immune responses to CMV and pathological processes during infection. This review discusses state-of-the-art knowledge of the immune response to CMV infection in tissues, with particular emphasis on several well-studied and most commonly affected organs.
Collapse
Affiliation(s)
- Andrea Mihalić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Jelena Železnjak
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Berislav Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stipan Jonjić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- Department of Biomedical Sciences, Croatian Academy of Sciences and Arts, Rijeka, Croatia
| | - Vanda Juranić Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| | - Ilija Brizić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia.
| |
Collapse
|
120
|
Meskini M, Zamani MS, Amanzadeh A, Bouzari S, Karimipoor M, Fuso A, Fateh A, Siadat SD. Epigenetic modulation of cytokine expression in Mycobacterium tuberculosis-infected monocyte derived-dendritic cells: Implications for tuberculosis diagnosis. Cytokine 2024; 181:156693. [PMID: 38986252 DOI: 10.1016/j.cyto.2024.156693] [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: 05/29/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND To delineate alterations in DNA methylation at high resolution within the genomic profile of monocyte-derived-dendritic cells (mo-DCs) in connection with Mycobacterium tuberculosis (MTB) infection, with particular emphasis on pro/ anti-inflammatory genes. METHODS In the context of this investigation, mo-DCs were infected by various active strains of MTB (Rifampicin-resistant [RIFR], H37Rv, multidrug-resistant [MDR], and extensively drug-resistant [XDR]). Subsequently, the pro/anti-inflammatory hub gene expression levels within the IL-6, IL-12, IFN-γ, IL-1β, TNF-α, and IL-10 pathways were evaluated employing real-time reverse transcription-polymerase chain reaction (RT-PCR). Additionally, the effects of MTB infection on mo-DC protein expression were examined through western blot analysis. The methylation status (%) of TNF-α and IL-10 was considered through Methylation Sensitive-High Resolution Melting (MS-HRM). RESULTS The results revealed an up-regulation of all pro-inflammatory genes among all groups, with TNF-α exhibiting the highest expression level. Conversely, the anti-inflammatory gene (IL-10) showed a down-regulated expression level. Furthermore, the DNA methylation status (%) of TNF-α decreased significantly among all the groups (P < 0.001), although there were no notable distinctions in the DNA methylation status (%) of IL-10 when compared to the control group (P > 0.05). CONCLUSION MTB infection induces DNA methylation changes in mo-DCs. The hypo-methylation of TNF-α may induce the up-regulation of this gene. This correlation revealed that the more resistant the MTB strain (XDR) is, the lower the methylation status (%) in the TNF-α gene.
Collapse
Affiliation(s)
- Maryam Meskini
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran; Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Saber Zamani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Amir Amanzadeh
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Saeid Bouzari
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Morteza Karimipoor
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran; Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Andrea Fuso
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
121
|
Bratosiewicz-Wąsik J, Wąsik TJ. Genetic variants of IL-10 promoter influence susceptibility to HIV-1 infection and disease progression in the Polish population: IL-10 polymorphisms and HIV-1. Hum Immunol 2024; 85:111086. [PMID: 39153369 DOI: 10.1016/j.humimm.2024.111086] [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: 05/16/2024] [Revised: 07/23/2024] [Accepted: 08/04/2024] [Indexed: 08/19/2024]
Abstract
The risk of HIV-1 infection and the rate of disease progression vary considerably among individuals and the genetic makeup of the host may be one of the possible reasons for this. We aimed to determine association of functional single nucleotide polymorphism (SNPs), -1082A/G (rs1800896), -819C/T (rs1800871), and -592C/A (rs1800872) in IL-10 gene, with the susceptibility to HIV-1 infection and clinical parameters expressed as a baseline CD4+ T cell count, CD8+ T cell count, and viral load. Therapy naïve HIV-1 infected individuals and HIV-1 seronegative controls from Poland were recruited for this study. Genotyping results revealed significantly higher frequency of -1082G/G genotype (28.1 % vs 16.1 %; p = 0.0019, OR=0.49) and -1082G allele (47.6 % vs 38.8 %; p = 0.0028, OR = 0.70) as well as lower frequency of -592 and -819 heterozygosity (45.0 % vs 34.4 %; p = 0.0266, OR = 1.47) in controls compared to seropositive subjects. High producing haplotype GCC was associated with increased risk of HIV-1 infection (p = 0.0018, OR = 1.52). Individuals possessing -592 and -819 minor allele had significantly higher CD8+ T cell count compared to the wild type allele carriers (p = 0.0303). Moreover, presence of -1082G allele was related with lower viral load as well as CD4+ and CD8+ T cells counts among patients infected with R5 HIV-1 variant. Thus, IL-10 gene promoter variants may be a risk factor for HIV-1 transmission and may modulate disease progression in the Polish population.
Collapse
Affiliation(s)
- Jolanta Bratosiewicz-Wąsik
- Department of Microbiology and Virology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, ul. Jagiellońska 4, Poland.
| | - Tomasz J Wąsik
- Department of Medical Microbiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, 40-752 Katowice, ul. Medyków 18, Poland.
| |
Collapse
|
122
|
Abreu MC, Conrad NL, Gonçalves VS, Leite FPL. Bacillus toyonensis amplifies the immunogenicity of an experimental recombinant tetanus vaccine in horses. J Equine Vet Sci 2024; 140:105135. [PMID: 38914241 DOI: 10.1016/j.jevs.2024.105135] [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: 11/02/2023] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
Probiotic microorganisms can stimulate an immune response and increase the efficiency of vaccines. For example, Bacillus toyonensis is a nonpathogenic, Gram-positive bacterium that has been used as a probiotic in animal supplementation. It induces immunomodulatory effects and increases the vaccine response in several species. This study aimed to evaluate the effect of B. toyonensis supplementation on the modulation of the immune response in horses vaccinated with recombinant Clostridium tetani toxin. Twenty horses were vaccinated twice, with an interval of 21 days between doses, and equally divided into two groups: the first group was supplemented orally for 42 days with feed containing viable spores of B. toyonensis (1 × 108) mixed with molasses (40 ml), starting 7 days before the first vaccination; the second (control) group received only feed mixed with molasses, starting 7 days before the first vaccination. Serum samples were collected to evaluate the humoral immune response using an in-house indirect enzyme-linked immunosorbent assay (ELISA), and peripheral blood mononuclear cells (PBMCs) were collected to evaluate cytokine transcription (qPCR). For the specific IgG-anti-rTENT titer, the supplemented group had ELISA values that were four times higher than those of the control group (p < 0.05). The supplemented group also showed higher ELISA values for the IgGa and IgGT sub-isotypes compared to the control group. In PBMCs stimulated with B. toyonensis, relative cytokine transcription of the supplemented group showed 15-, 8-, 7-, and 6-fold increases for IL1, TNFα, IL10 and IL4, respectively. When stimulated with a vaccine antigen, the supplemented group showed 1.6-, 1.8-, and 0.5-fold increases in IL1, TNFα, and IL4, respectively, compared to the control group. Horses supplemented with B. toyonensis had a significantly improved vaccine immune response compared to those in the control group, which suggests a promising approach for improving vaccine efficacy with probiotics.
Collapse
Affiliation(s)
- Mayara Caetano Abreu
- Departament of Veterinary Medicine, Federal University of Pelotas, UFPel, Capão do Leão, Rio Grande do Sul, Brazil
| | - Neida Lucia Conrad
- Center for Technological Development, Biotecnology, Federal University of Pelotas, UFPel, Capão do Leão, Rio Grande do Sul, Brazil
| | - Vitória Sequeira Gonçalves
- Center for Technological Development, Biotecnology, Federal University of Pelotas, UFPel, Capão do Leão, Rio Grande do Sul, Brazil
| | - Fábio Pereira Leivas Leite
- Departament of Veterinary Medicine, Federal University of Pelotas, UFPel, Capão do Leão, Rio Grande do Sul, Brazil; Center for Technological Development, Biotecnology, Federal University of Pelotas, UFPel, Capão do Leão, Rio Grande do Sul, Brazil.
| |
Collapse
|
123
|
Mani V, Arfeen M. Betahistine's Neuroprotective Actions against Lipopolysaccharide-Induced Neurotoxicity: Insights from Experimental and Computational Studies. Brain Sci 2024; 14:876. [PMID: 39335372 PMCID: PMC11430358 DOI: 10.3390/brainsci14090876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Histamine H3 receptor (H3R) antagonists, such as betahistine (BHTE), have shown significant potential in treating central nervous system (CNS) disorders due to their neuroprotective properties. This study investigated BHTE's effects on lipopolysaccharide (LPS)-induced neurotoxicity, which is associated with neuroinflammation and neurodegeneration. Rats were divided into groups and pre-treated with BHTE (5 or 10 mg/kg, p.o.) for 30 days, followed by LPS administration (1 mg/kg, i.p.) for 4 consecutive days to induce neurotoxicity. LPS exposure resulted in cognitive impairment, as evidenced by performance deficits in maze tests, and a significant reduction in brain acetylcholine (ACh) levels. Additionally, LPS led to increased neuroinflammation, oxidative stress, mitochondrial dysfunction, and apoptosis. Pre-treatment with BHTE effectively counteracted these effects, improving cognitive performance and restoring ACh levels. BHTE significantly reduced LPS-induced increases in pro-inflammatory markers (COX-2, TNF-α, and IL-6) while enhancing anti-inflammatory cytokines (IL-10 and TGF-β1). Furthermore, BHTE improved mitochondrial function by increasing enzyme levels (MRCC-I, II, and IV) and boosted anti-apoptotic (Bcl-2) and antioxidant defenses (GSH and catalase). BHTE also reduced apoptosis markers, including pro-apoptotic protein caspase-3, and oxidative stress marker malondialdehyde (MDA). Molecular modeling studies revealed that BHTE effectively binds to key enzymes involved in neuroinflammation and apoptosis (AChE, COX-2, and caspase-3), with binding free energies between 4 and 5 kcal/mol, interacting with critical residues. These findings underscore BHTE's multifaceted neuroprotective effects against LPS-induced neurotoxicity, offering potential therapeutic avenues for managing neuroinflammation and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Vasudevan Mani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Minhajul Arfeen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia;
| |
Collapse
|
124
|
Oğuzoğlu TÇ, Hanifehnezhad A, Khabbazi SD, Karayel-Hacıoğlu İ, Kaynarcalıdan O, Fırat Z, Filazi N, Erdem-Şahinkesen E, Gül B, Karabulut MC, Koba E, Adıgüzel E, Şenlik Eİ, Korkulu E, Demirden C, Şahinkesen İ, Ceylan A, Muratoğlu H, Vural S, Demirbağ Z, Özkul A. Immunogenicity and Protective Efficacy of Baculovirus-Expressed SARS-CoV-2 Envelope Protein in Mice as a Universal Vaccine Candidate. Vaccines (Basel) 2024; 12:977. [PMID: 39340009 PMCID: PMC11435448 DOI: 10.3390/vaccines12090977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 09/30/2024] Open
Abstract
The envelope (env) protein of SARS-CoV-2, a pivotal component of the viral architecture, plays a multifaceted role in viral assembly, replication, pathogenesis, and ion channel activity. These features make it a significant target for understanding virus-host interactions and developing vaccines to combat COVID-19. Recent structural studies provide valuable insights into the conformational dynamics and membrane topology of the SARS-CoV-2 env protein, shedding light on its functional mechanisms. The strong homology and highly conserved structure of the SARS-CoV-2 env protein shape its immunogenicity and functional characteristics. This study examines the ability of the recombinant SARS-CoV-2 env protein to stimulate an immune response. In this study, recombinant envelope proteins were produced using the baculovirus expression system, and their potential efficacy was evaluated in both in vivo and in vitro models. Our results reveal that the env protein of SARS-CoV-2 stimulates humoral and cellular responses and highlight its potential as a promising vaccine candidate for combating the ongoing pandemic.
Collapse
Affiliation(s)
- Tuba Çiğdem Oğuzoğlu
- Department of Virology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
| | - Alireza Hanifehnezhad
- Department of Virology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
| | - Saber Delpasand Khabbazi
- Department of Agriculture and Food, Institute of Hemp Research, Yozgat Bozok University, Yozgat 66900, Türkiye
| | - İlke Karayel-Hacıoğlu
- Department of Virology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
| | - Onur Kaynarcalıdan
- Institute for Virology, Düsseldorf University Hospital, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Zehra Fırat
- Biotechnology Institute, Ankara University, Ankara 06560, Türkiye
| | - Nazlıcan Filazi
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
- Department of Virology, Faculty of Veterinary Medicine, Hatay Mustafa Kemal University, Hatay 31040, Türkiye
| | - Eda Erdem-Şahinkesen
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
- Department of Vaccine Technology, Vaccine Institute, Hacettepe University, Ankara 06100, Türkiye
| | - Buket Gül
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
| | - Muhammed Cesim Karabulut
- Department of Virology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
| | - Enes Koba
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
| | - Ece Adıgüzel
- Republic of Türkiye Ministry of Agriculture and Forestry, Atkaracalar District Directorate, Çankırı 18310, Türkiye
| | - Elif İrem Şenlik
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
| | - Emrah Korkulu
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
- Department of Virology, Faculty of Veterinary Medicine, Kafkas University, Kars 36000, Türkiye
| | - Cansu Demirden
- Graduate School of Health Sciences, Ankara University, Ankara 06560, Türkiye
- Republic of Türkiye Ministry of Agriculture and Forestry, East Anatolian Agricultural Research Institute, Erzurum 25090, Türkiye
| | - İlker Şahinkesen
- Diagen Biotechnological Systems Health Services and Automation Industry, Ankara 06070, Türkiye
| | - Ahmet Ceylan
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
| | - Hacer Muratoğlu
- Department of Molecular Biology and Genetics, Faculty of Sciences, Karadeniz Technical University, Trabzon 61080, Türkiye
| | - Sevil Vural
- Department of Pathology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
| | - Zihni Demirbağ
- Department of Biology, Faculty of Sciences, Karadeniz Technical University, Trabzon 61080, Türkiye
| | - Aykut Özkul
- Department of Virology, Faculty of Veterinary Medicine, Ankara University, Ankara 06070, Türkiye
| |
Collapse
|
125
|
Ivanovski F, Meško M, Lebar T, Rupnik M, Lainšček D, Gradišek M, Jerala R, Benčina M. Ultrasound-mediated spatial and temporal control of engineered cells in vivo. Nat Commun 2024; 15:7369. [PMID: 39191796 DOI: 10.1038/s41467-024-51620-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Remote regulation of cells in deep tissue remains a significant challenge. Low-intensity pulsed ultrasound offers promise for in vivo therapies due to its non-invasive nature and precise control. This study uses pulsed ultrasound to control calcium influx in mammalian cells and engineers a therapeutic cellular device responsive to acoustic stimulation in deep tissue without overexpressing calcium channels or gas vesicles. Pulsed ultrasound parameters are established to induce calcium influx in HEK293 cells. Additionally, cells are engineered to express a designed calcium-responsive transcription factor controlling the expression of a selected therapeutic gene, constituting a therapeutic cellular device. The engineered sonogenetic system's functionality is demonstrated in vivo in mice, where an implanted anti-inflammatory cytokine-producing cellular device effectively alleviates acute colitis, as shown by improved colonic morphology and histopathology. This approach provides a powerful tool for precise, localized control of engineered cells in deep tissue, showcasing its potential for targeted therapeutic delivery.
Collapse
Affiliation(s)
- Filip Ivanovski
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia
- Interfaculty Doctoral Study of Biomedicine, University of Ljubljana, Vrazov trg 2, Ljubljana, Slovenia
| | - Maja Meško
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia
| | - Tina Lebar
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia
| | - Marko Rupnik
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia
| | - Miha Gradišek
- Faculty of Electrical Engineering, University of Ljubljana, Tržaška c. 25, Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia.
- CTGCT, Centre of Technology of Gene and Cell Therapy, Hajdrihova 19, Ljubljana, Slovenia.
| | - Mojca Benčina
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, Slovenia.
- CTGCT, Centre of Technology of Gene and Cell Therapy, Hajdrihova 19, Ljubljana, Slovenia.
- University of Ljubljana, Kongresni trg 12, 1000, Ljubljana, Slovenia.
| |
Collapse
|
126
|
Rajput Y, Neral A, Sherwani N, Jain V, Sahu M, Paikra F, Kushwaha A, Sahu A, Lodhi H, Sundrani O, Panda RK, Jain V, Shammas MA, Pal J. A novel metric-based approach of scoring early host immune response from oro-nasopharyngeal swabs predicts COVID-19 outcome. Sci Rep 2024; 14:19510. [PMID: 39174586 PMCID: PMC11341902 DOI: 10.1038/s41598-024-70161-8] [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: 02/05/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
Unpredictable fatal outcome of COVID-19 is attributed to dysregulated inflammation. Impaired early adaptive immune response leads to late-stage inflammatory outcome. The purpose of this study was to develop biomarkers for early detection of host immune impairment at first diagnosis from leftover RNA samples, which may in turn identify high risk patients. Leftover RNA samples of COVID-19 patients at first diagnosis were stored. Following prospective follow-up, the samples were shorted and categorized into outcome groups. Impaired adaptive T cell response (severity score) and Impaired IL-10 response (undetectable IL-10 in the presence of high expression of a representative interferon response gene) were determined by RT-PCR based assay. We demonstrate that a T cell response based 'severity score' comprising rational combination of Ct values of a target genes' signature can predict high risk noncomorbid potentially critical COVID-19 patients with a sensitivity of 91% (95% CI 58.7-99.8) and specificity of 92.6% (95% CI 75.7-99) (AUC:0.88). Although inclusion of comorbid patients reduced sensitivity to 77% (95% CI 54.6-92.2), the specificity was still 94% (95% CI 79.8-99.3) (AUC:0.82). The same for 'impaired IL-10 response' were little lower to predict high risk noncomorbid patients 64.2% (95% CI 35.1-87.2) and 82% (95% CI 65.5-93.2) respectively. Inclusion of comorbid patients drastically reduce sensitivity and specificity51.6% (95% CI 33.1-69.8) and 80.5% (95% CI 64.0-91.8) respectively. As best of our knowledge this is the first demonstration of a metric-based approach showing the 'severity score' as an indicator of early adoptive immune response, could be used as predictor of severe COVID-19 outcome at the time of first diagnosis using the same leftover swab RNA. The work flow could reduce expenditure and reporting time of the prognostic test for an earliest clinical decision ensuring possibility of early rational management.
Collapse
Affiliation(s)
- Yogita Rajput
- Multidisciplinary Research Units (MRU), Pt. J.N.M. Medical College, Raipur, Chhattisgarh, 492001, India
| | - Arvind Neral
- Department of Microbiology, Pt. J.N.M. Medical College, Raipur, C.G., India
- Department of Pathology, Pt. J.N.M. Medical College, Raipur, C.G, India
| | - Nikita Sherwani
- Department of Microbiology, Pt. J.N.M. Medical College, Raipur, C.G., India
| | - Vijaylakshmi Jain
- Department of Microbiology, Pt. J.N.M. Medical College, Raipur, C.G., India
| | - Malti Sahu
- Multidisciplinary Research Units (MRU), Pt. J.N.M. Medical College, Raipur, Chhattisgarh, 492001, India
| | - Fulsay Paikra
- Multidisciplinary Research Units (MRU), Pt. J.N.M. Medical College, Raipur, Chhattisgarh, 492001, India
| | - Aarti Kushwaha
- Multidisciplinary Research Units (MRU), Pt. J.N.M. Medical College, Raipur, Chhattisgarh, 492001, India
| | - Aparna Sahu
- Department of Microbiology, Pt. J.N.M. Medical College, Raipur, C.G., India
| | - Heeramani Lodhi
- Department of Anaesthesia and Pain Management, Pt. J.N.M. Medical College, Raipur, C.G, India
| | - Omprakash Sundrani
- Department of Critical Care Medicine, Pt. J.N.M. Medical College, Raipur, C.G., India
| | - Ravindra Kumar Panda
- Department of Respiratory Medicine, Pt. J.N.M. Medical College, Raipur, C.G., India
| | - Vinit Jain
- Superintendent (past), Dr. BRAM Hospital Raipur CG and Department of Orthopaedics, Pt. J.N.M. Medical College, Raipur, C.G., India
| | - Masood A Shammas
- Department of Adult Oncology, Harvard (Dana Farber) Cancer Institute and VA Health Care System, Boston, MA, USA
| | - Jagannath Pal
- Multidisciplinary Research Units (MRU), Pt. J.N.M. Medical College, Raipur, Chhattisgarh, 492001, India.
| |
Collapse
|
127
|
Chin YT, Tsai CL, Ma HH, Cheng DC, Tsai CW, Wang YC, Shih HY, Chang SY, Gu J, Chang WS, Bau DT. Impacts of Interleukin-10 Promoter Genotypes on Prostate Cancer. Life (Basel) 2024; 14:1035. [PMID: 39202777 PMCID: PMC11355935 DOI: 10.3390/life14081035] [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: 07/22/2024] [Revised: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Prostate cancer (PCa) is a multifactorial disease influenced by genetic, environmental, and immunological factors. Genetic polymorphisms in the interleukin-10 (IL-10) gene have been implicated in PCa susceptibility, development, and progression. This study aims to assess the contributions of three IL-10 promoter single nucleotide polymorphisms (SNPs), A-1082G (rs1800896), T-819C (rs3021097), and A-592C (rs1800872), to the risk of PCa in Taiwan. The three IL-10 genotypes were determined using PCR-RFLP methodology and were evaluated for their contributions to PCa risk among 218 PCa patients and 436 non-PCa controls. None of the three IL-10 SNPs were significantly associated with the risks of PCa (p all > 0.05) in the overall analyses. However, the GG at rs1800896 combined with smoking behavior was found to significantly increase the risk of PCa by 3.90-fold (95% confidence interval [95% CI] = 1.28-11.89, p = 0.0231). In addition, the rs1800896 AG and GGs were found to be correlated with the late stages of PCa (odds ratio [OR] = 1.90 and 6.42, 95% CI = 1.05-3.45 and 2.30-17.89, p = 0.0452 and 0.0003, respectively). The IL-10 promoter SNP, A-1082G (rs1800896), might be a risk factor for PCa development among smokers and those at late stages of the disease. These findings should be validated in larger and more diverse populations.
Collapse
Affiliation(s)
- Yu-Ting Chin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung 404327, Taiwan
| | - Chung-Lin Tsai
- Division of Cardiac and Vascular Surgery, Cardiovascular Center, Taichung Veterans General Hospital, Taichung 407219, Taiwan
| | - Hung-Huan Ma
- Division of Nephrology, Department of Internal Medicine, Taichung Tzu Chi Hospital, Taichung 427003, Taiwan
| | - Da-Chuan Cheng
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung 404333, Taiwan
| | - Chia-Wen Tsai
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung 404327, Taiwan
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Yun-Chi Wang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung 404327, Taiwan
| | - Hou-Yu Shih
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung 404327, Taiwan
| | - Shu-Yu Chang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung 404327, Taiwan
- Department of Nephrology, Chang-Hua Hospital, Ministry of Health and Welfare, Changhua 51341, Taiwan
| | - Jian Gu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Wen-Shin Chang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung 404327, Taiwan
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Da-Tian Bau
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung 404327, Taiwan
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 413305, Taiwan
| |
Collapse
|
128
|
Hoffmann SW, Schierbauer J, Zimmermann P, Voit T, Grothoff A, Wachsmuth NB, Rössler A, Niedrist T, Lackner HK, Moser O. Effects of Interrupting Prolonged Sitting with Light-Intensity Physical Activity on Inflammatory and Cardiometabolic Risk Markers in Young Adults with Overweight and Obesity: Secondary Outcome Analyses of the SED-ACT Randomized Controlled Crossover Trial. Biomolecules 2024; 14:1029. [PMID: 39199416 PMCID: PMC11352707 DOI: 10.3390/biom14081029] [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: 07/30/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/01/2024] Open
Abstract
Sedentary behavior (SB) is an essential risk factor for obesity, cardiovascular disease, and type 2 diabetes. Though certain levels of physical activity (PA) may attenuate the detrimental effects of SB, the inflammatory and cardiometabolic responses involved are still not fully understood. The focus of this secondary outcome analysis was to describe how light-intensity PA snacks (LIPASs, alternate sitting and standing, walking or standing continuously) compared with uninterrupted prolonged sitting affect inflammatory and cardiometabolic risk markers. Seventeen young adults with overweight and obesity participated in this study (eight females, 23.4 ± 3.3 years, body mass index (BMI) 29.7 ± 3.8 kg/m2, glycated hemoglobin A1C (HbA1c) 5.4 ± 0.3%, body fat 31.8 ± 8.2%). Participants were randomly assigned to the following conditions which were tested during an 8 h simulated workday: uninterrupted prolonged sitting (SIT), alternate sitting and standing (SIT-STAND, 2.5 h total standing time), continuous standing (STAND), and continuous walking (1.6 km/h; WALK). Each condition also included a standardized non-relativized breakfast and lunch. Venous blood samples were obtained in a fasted state at baseline (T0), 1 h after lunch (T1) and 8 h after baseline (T2). Inflammatory and cardiometabolic risk markers included interleukin-6 (IL-6), c-reactive protein (CRP), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), triglycerides (TGs), visceral fat area (VFA), triglyceride-glucose (TyG) index, two lipid ratio measures, TG/HDL-C and TC/HDL-C, albumin, amylase (pancreatic), total protein, uric acid, and urea. We found significant changes in a broad range of certain inflammatory and cardiometabolic risk markers during the intervention phase for IL-6 (p = 0.014), TG (p = 0.012), TC (p = 0.017), HDL-C (p = 0.020), LDL-C (p = 0.021), albumin (p = 0.003), total protein (p = 0.021), and uric acid (p = 0.040) in favor of light-intensity walking compared with uninterrupted prolonged sitting, alternate sitting and standing, and continuous standing. We found no significant changes in CRP (p = 0.529), creatinine (p = 0.199), TyG (p = 0.331), and the lipid ratios TG/HDL-C (p = 0.793) and TC/HDL-C (p = 0.221) in response to the PA snack. During a simulated 8 h work environment replacement and interruption of prolonged sitting with light-intensity walking, significant positive effects on certain inflammatory and cardiometabolic risk markers were found in young adults with overweight and obesity.
Collapse
Affiliation(s)
- Sascha W. Hoffmann
- Division of Theory and Practice of Sports and Fields of Physical Activity, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany
| | - Janis Schierbauer
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (J.S.); (P.Z.); (T.V.); (A.G.); (N.B.W.)
| | - Paul Zimmermann
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (J.S.); (P.Z.); (T.V.); (A.G.); (N.B.W.)
| | - Thomas Voit
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (J.S.); (P.Z.); (T.V.); (A.G.); (N.B.W.)
| | - Auguste Grothoff
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (J.S.); (P.Z.); (T.V.); (A.G.); (N.B.W.)
| | - Nadine B. Wachsmuth
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (J.S.); (P.Z.); (T.V.); (A.G.); (N.B.W.)
| | - Andreas Rössler
- Department of Physiology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria; (A.R.); (H.K.L.)
| | - Tobias Niedrist
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8010 Graz, Austria;
| | - Helmut K. Lackner
- Department of Physiology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria; (A.R.); (H.K.L.)
| | - Othmar Moser
- Division of Exercise Physiology and Metabolism, BaySpo—Bayreuth Center of Sport Science, University of Bayreuth, 95440 Bayreuth, Germany; (J.S.); (P.Z.); (T.V.); (A.G.); (N.B.W.)
- Interdisciplinary Metabolic Medicine Trials Unit, Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, 8010 Graz, Austria
| |
Collapse
|
129
|
Gilio L, Fresegna D, Stampanoni Bassi M, Musella A, De Vito F, Balletta S, Sanna K, Caioli S, Pavone L, Galifi G, Simonelli I, Guadalupi L, Vanni V, Buttari F, Dolcetti E, Bruno A, Azzolini F, Borrelli A, Fantozzi R, Finardi A, Furlan R, Centonze D, Mandolesi G. Interleukin-10 contrasts inflammatory synaptopathy and central neurodegenerative damage in multiple sclerosis. Front Mol Neurosci 2024; 17:1430080. [PMID: 39169949 PMCID: PMC11338018 DOI: 10.3389/fnmol.2024.1430080] [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: 05/09/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024] Open
Abstract
Proinflammatory cytokines are implicated in promoting neurodegeneration in multiple sclerosis (MS) by affecting excitatory and inhibitory transmission at central synapses. Conversely, the synaptic effects of anti-inflammatory molecules remain underexplored, despite their potential neuroprotective properties and their presence in the cerebrospinal fluid (CSF) of patients. In a study involving 184 newly diagnosed relapsing-remitting (RR)-MS patients, we investigated whether CSF levels of the anti-inflammatory interleukin (IL)-10 were linked to disease severity and neurodegeneration measures. Additionally, we examined IL-10 impact on synaptic transmission in striatal medium spiny neurons and its role in counteracting inflammatory synaptopathy induced by IL-1β in female C57BL/6 mice with experimental autoimmune encephalomyelitis (EAE). Our findings revealed a significant positive correlation between IL-10 CSF levels and changes in EDSS (Expanded Disability Status Scale) scores one year after MS diagnosis. Moreover, IL-10 levels in the CSF were positively correlated with volumes of specific subcortical brain structures, such as the nucleus caudate. In both MS patients' CSF and EAE mice striatum, IL-10 and IL-1β expressions were upregulated, suggesting possible antagonistic effects of these cytokines. Notably, IL-10 exhibited the ability to decrease glutamate transmission, increase GABA transmission in the striatum, and reverse IL-1β-induced abnormal synaptic transmission in EAE. In conclusion, our data suggest that IL-10 exerts direct neuroprotective effects in MS patients by modulating both excitatory and inhibitory transmission and attenuating IL-1β-induced inflammatory synaptopathy. These findings underscore the potential therapeutic significance of IL-10 in mitigating neurodegeneration in MS.
Collapse
Affiliation(s)
- Luana Gilio
- Neurology Unit, IRCCS Neuromed, Pozzilli, Italy
- Faculty of Psychology, Uninettuno Telematic International University, Rome, Italy
| | - Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Roma, Italy
| | | | | | - Krizia Sanna
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Giovanni Galifi
- Neurology Unit, IRCCS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ilaria Simonelli
- Clinical Trial Centre Isola Tiberina-Gemelli Isola, Rome, Italy
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Livia Guadalupi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Valentina Vanni
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
| | - Fabio Buttari
- Neurology Unit, IRCCS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ettore Dolcetti
- Neurology Unit, IRCCS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Antonio Bruno
- Neurology Unit, IRCCS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | | | | | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- IRCCS San Raffaele Scientific Institute, University Vita-Salute San Raffaele, Milan, Italy
| | - Diego Centonze
- Neurology Unit, IRCCS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Roma, Italy
| |
Collapse
|
130
|
Lee D, Jo MG, Min KY, Choi MY, Kim YM, Kim HS, Choi WS. IL-10 + regulatory B cells mitigate atopic dermatitis by suppressing eosinophil activation. Sci Rep 2024; 14:18164. [PMID: 39107352 PMCID: PMC11303538 DOI: 10.1038/s41598-024-68660-9] [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: 05/29/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Atopic dermatitis (AD) presents significant therapeutic challenges due to its poorly understood etiology. Eosinophilia, a hallmark of allergic inflammation, is implicated in AD pathogenesis. Interleukin-10 (IL-10)-producing regulatory B (Breg) cells exhibit potent anti-inflammatory effects. However, their role in controlling AD-related eosinophilia is not well understood. To investigate the impact of eosinophils on AD, we employed IL-5Rα-deficient (Il5ra-/-) mice, which lack functional eosinophils. Induction of AD in these mice resulted in attenuated disease symptoms, underscoring the critical role of eosinophils in AD development. Additionally, the adoptive transfer of purified Breg cells into mice with AD significantly alleviated disease severity. Mechanistic studies revealed that IL-10 produced by Breg cells directly inhibits eosinophil activation and infiltration into the skin. In vitro experiments further confirmed that Breg cells inhibited eosinophil peroxidase secretion in an IL-10-dependent manner. Our collective findings demonstrate that IL-10 from Breg cells alleviates AD by suppressing eosinophil activation and tissue infiltration. This study elucidates a novel regulatory mechanism of Breg cells, providing a foundation for future Breg-mediated therapeutic strategies for AD.
Collapse
Affiliation(s)
- Dajeong Lee
- School of Medicine, Konkuk University, Chungju, 27478, Korea
| | - Min Geun Jo
- School of Medicine, Konkuk University, Chungju, 27478, Korea
| | - Keun Young Min
- School of Medicine, Konkuk University, Chungju, 27478, Korea
| | - Min Yeong Choi
- School of Medicine, Konkuk University, Chungju, 27478, Korea
| | - Young Mi Kim
- College of Pharmacy, Duksung Women's University, Seoul, 01369, Korea
| | - Hyuk Soon Kim
- Department of Biomedical Sciences, College of Natural Science and Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315, Korea.
| | - Wahn Soo Choi
- School of Medicine, Konkuk University, Chungju, 27478, Korea.
- Institute of Biomedical Sciences & Technology, Konkuk University, Seoul, 05029, Korea.
| |
Collapse
|
131
|
Costa GL, Sautto GA. Exploring T-Cell Immunity to Hepatitis C Virus: Insights from Different Vaccine and Antigen Presentation Strategies. Vaccines (Basel) 2024; 12:890. [PMID: 39204016 PMCID: PMC11359689 DOI: 10.3390/vaccines12080890] [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: 06/21/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
The hepatitis C virus (HCV) is responsible for approximately 50 million infections worldwide. Effective drug treatments while available face access barriers, and vaccine development is hampered by viral hypervariability and immune evasion mechanisms. The CD4+ and CD8+ T-cell responses targeting HCV non-structural (NS) proteins have shown a role in the viral clearance. In this paper, we reviewed the studies exploring the relationship between HCV structural and NS proteins and their effects in contributing to the elicitation of an effective T-cell immune response. The use of different vaccine platforms, such as viral vectors and virus-like particles, underscores their versability and efficacy for vaccine development. Diverse HCV antigens demonstrated immunogenicity, eliciting a robust immune response, positioning them as promising vaccine candidates for protein/peptide-, DNA-, or RNA-based vaccines. Moreover, adjuvant selection plays a pivotal role in modulating the immune response. This review emphasizes the importance of HCV proteins and vaccination strategies in vaccine development. In particular, the NS proteins are the main focus, given their pivotal role in T-cell-mediated immunity and their sequence conservation, making them valuable vaccine targets.
Collapse
Affiliation(s)
| | - Giuseppe A. Sautto
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA;
| |
Collapse
|
132
|
Yehia SA, Badr AM, Bashtar AR, Ibrahim MAA, Mousa MR, Mostafa NA. Immune response, oxidative stress, and histological changes of Wistar rats after being administered with Parascaris equorum antigen. Sci Rep 2024; 14:18069. [PMID: 39103392 PMCID: PMC11300452 DOI: 10.1038/s41598-024-67788-y] [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: 07/16/2024] [Indexed: 08/07/2024] Open
Abstract
Worldwide, particularly in developing nations, helminth infections are the leading causes of livestock illness and mortality. Parascaris (P.) equorum, a parasitic worm from the Ascarididae family, significantly impacts the production, health, and working performance of equines. This study aimed to investigate the impact of intraperitoneal sensitization of P. equorum on the immune system, oxidative stress, and histology in Wistar rats. After acclimatization for 7 days, we divided the rats into five groups, each consisting of six rats. Group I, serving as the control, was administered distilled water, followed by groups II (day 7), III (day 14), IV (day 21), and V (day 33). The rats were euthanized every day mentioned (Days 7-33). On day 0, a dosage of 1ml/100 gm rat (containing 500 μg/ml protein content) emulsified crude antigen extract with an incomplete Freund's adjuvant (1:1 volume), followed by a second dose of the same antigen concentration on day 7. To assess the allergenicity of this nematode, we measured a whole blood profile, serum levels of IFN-γ, IL-5, IL-10, IL-13, and IL-33, total immunoglobulins IgE and IgG, and oxidative stress markers. Also, we examined histological changes in the liver, kidney, and spleen. The results showed that values of total leukocyte count, granulocytes, monocytes, and lymphocytes were significantly (P < 0.05) increased on day 14 post-infection relative to other days of investigation. It was found that the levels of total immunoglobulins (IgE and IgG) and cytokines (INF-γ, IL-5, IL-13, and IL-33) on days 14 and 21 were significantly higher than in the control group. At all periods of the experiment, the injected group exhibited significantly higher concentrations of MDA and NO compared to the control group (P < 0.05). Conversely, GSH and CAT levels (P < 0.05) dropped significantly on days 7, 14, and 21. Different rat tissues showed alterations. Ultimately, this study described the detrimental effects of P. equorum crude antigen administration on the immune system, oxidative states, and histological changes of Wistar rats at various intervals.
Collapse
Affiliation(s)
- Salma Adel Yehia
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | | | | | | | - Mohamed Refat Mousa
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | | |
Collapse
|
133
|
Nagarajan A, Scoggin K, Adams LG, Threadgill D, Andrews-Polymenis H. Identification of a genetic region linked to tolerance to MRSA infection using Collaborative Cross mice. PLoS Genet 2024; 20:e1011378. [PMID: 39178306 PMCID: PMC11407622 DOI: 10.1371/journal.pgen.1011378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 09/17/2024] [Accepted: 07/29/2024] [Indexed: 08/25/2024] Open
Abstract
Staphylococcus aureus (S. aureus) colonizes humans asymptomatically but can also cause opportunistic infections, ranging from mild skin infections to severe life-threatening conditions. Resistance and tolerance are two ways a host can survive an infection. Resistance is limiting the pathogen burden, while tolerance is limiting the health impact of a given pathogen burden. In previous work, we established that collaborative cross (CC) mouse line CC061 is highly susceptible to Methicillin-resistant S. aureus infection (MRSA, USA300), while CC024 is tolerant. To identify host genes involved in tolerance after S. aureus infection, we crossed CC061 mice and CC024 mice to generate F1 and F2 populations. Survival after MRSA infection in the F1 and F2 generations was 65% and 55% and followed a complex dominant inheritance pattern for the CC024 increased survival phenotype. Colonization in F2 animals was more extreme than in their parents, suggesting successful segregation of genetic factors. We identified a Quantitative Trait Locus (QTL) peak on chromosome 7 for survival and weight change after infection. In this QTL, the WSB/EiJ (WSB) allele was present in CC024 mice and contributed to their MRSA tolerant phenotype. Two genes, C5ar1 and C5ar2, have high-impact variants in this region. C5ar1 and C5ar2 are receptors for the complement factor C5a, an anaphylatoxin that can trigger a massive immune response by binding to these receptors. We hypothesize that C5a may have altered binding to variant receptors in CC024 mice, reducing damage caused by the cytokine storm and resulting in the ability to tolerate a higher pathogen burden and longer survival.
Collapse
Affiliation(s)
- Aravindh Nagarajan
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| | - Kristin Scoggin
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| | - L Garry Adams
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - David Threadgill
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas, United States of America
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, Texas, United States of America
- Department of Biochemistry & Biophysics and Department of Nutrition, Texas A&M University, College Station, Texas, United States of America
| | - Helene Andrews-Polymenis
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
134
|
Opgenorth J, Abeyta MA, Goetz BM, Rodriguez-Jimenez S, Freestone AD, Rhoads RP, McMillan RP, McGill JL, Baumgard LH. Intramammary lipopolysaccharide challenge in early- versus mid-lactation dairy cattle: Immune, production, and metabolic responses. J Dairy Sci 2024; 107:6252-6267. [PMID: 38460880 DOI: 10.3168/jds.2023-24488] [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: 11/29/2023] [Accepted: 02/06/2024] [Indexed: 03/11/2024]
Abstract
Study objectives were to compare the immune response, metabolism, and production following intramammary LPS (IMM LPS) administration in early and mid-lactation cows. Early (E-LPS; n = 11; 20 ± 4 DIM) and mid- (M-LPS; n = 10; 155 ± 40 DIM) lactation cows were enrolled in an experiment consisting of 2 periods (P). During P1 (5 d) cows were fed ad libitum and baseline data were collected, including liver and muscle biopsies. At the beginning of P2 (3 d) cows received 10 mL of sterile saline containing 10 µg of LPS from Escherichia coli O111:B4/mL into the left rear quarter of the mammary gland, and liver and muscle biopsies were collected at 12 h after LPS. Tissues were analyzed for metabolic flexibility, which measures substrate switching capacity from pyruvic acid to palmitic acid oxidation. Data were analyzed with the MIXED procedure in SAS 9.4. Rectal temperature was assessed hourly for the first 12 h after LPS and every 6 h thereafter for the remainder of P2. All cows developed a febrile response following LPS, but E-LPS had a more intense fever than M-LPS cows (0.7°C at 5 h after LPS). Blood samples were collected at 0, 3, 6, 9, 12, 24, 36, 48, and 72 h after LPS for analysis of systemic inflammation and metabolism parameters. Total serum Ca decreased after LPS (26% at 6 h nadir) but did not differ by lactation stage (LS). Circulating neutrophils decreased, then increased after LPS in both LS, but E-LPS had exaggerated neutrophilia (56% from 12 to 48 h) compared with M-LPS. Haptoglobin increased after LPS (15-fold) but did not differ by LS. Many circulating cytokines were increased after LPS, and IL-6, IL-10, TNF-α, MCP-1, and IP-10 were further augmented in E-LPS compared with M-LPS cows. Relative to P1, all cows had reduced milk yield (26%) and DMI (14%) on d 1 that did not differ by LS. Somatic cell score increased rapidly in response to LPS regardless of LS and gradually decreased from 18 h onwards. Milk component yields decreased after LPS. However, E-LPS had increased fat (11%) and tended to have increased lactose (8%) yield compared with M-LPS cows throughout P2. Circulating glucose was not affected by LPS. Nonesterified fatty acids (NEFA) decreased in E-LPS (29%) but not M-LPS cows. β-Hydroxybutyrate slightly increased (14%) over time after LPS regardless of LS. Insulin increased after LPS in all cows, but E-LPS had blunted hyperinsulinemia (52%) compared with M-LPS cows. Blood urea nitrogen increased after LPS, and the relative change in BUN was elevated in E-LPS cows compared with M-LPS cows (36% and 13%, respectively, from 9 to 24 h). During P1, metabolic flexibility was increased in liver and muscle in early lactating cows compared with mid-lactation cows, but 12 h after LPS, metabolic flexibility was reduced and did not differ by LS. In conclusion, IMM LPS caused severe immune activation, and E-LPS cows had a more intense inflammatory response compared with M-LPS cows, but the effects on milk synthesis was similar between LS. Some parameters of the E-LPS metabolic profile suggest continuation of metabolic adjustments associated with early lactation to support both a robust immune system and milk synthesis.
Collapse
Affiliation(s)
- J Opgenorth
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - M A Abeyta
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - B M Goetz
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | | | - A D Freestone
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - R P Rhoads
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060
| | - R P McMillan
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060
| | - J L McGill
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, IA 50011
| | - L H Baumgard
- Department of Animal Science, Iowa State University, Ames, IA 50011.
| |
Collapse
|
135
|
Opgenorth J, Mayorga EJ, Abeyta MA, Goetz BM, Rodriguez-Jimenez S, Freestone AD, McGill JL, Baumgard LH. Intravenous lipopolysaccharide challenge in early- versus mid-lactation dairy cattle. I: The immune and inflammatory responses. J Dairy Sci 2024; 107:6225-6239. [PMID: 38428491 DOI: 10.3168/jds.2023-24350] [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: 10/25/2023] [Accepted: 02/02/2024] [Indexed: 03/03/2024]
Abstract
Cows in early lactation (EL) are purportedly immune suppressed, which renders them more susceptible to disease. Thus, the study objective was to compare key biomarkers of immune activation from i.v. LPS between EL and mid-lactation (ML) cows. Multiparous EL (20 ± 2 DIM; n = 11) and ML (131 ± 31 DIM; n = 12) cows were enrolled in a 2 × 2 factorial design and assigned to 1 of 2 treatments by lactation stage (LS): (1) EL (EL-LPS; n = 6) or ML (ML-LPS; n = 6) cows administered a single LPS bolus from Escherichia coli O55:B5 (0.09 µg/kg of BW), or (2) pair-fed (PF) EL (EL-PF; n = 5) or ML (ML-PF; n = 6) cows administered i.v. saline. After LPS administration, cows were intensely evaluated for 3 d to analyze their response and recovery to LPS. Rectal temperature increased in LPS relative to PF cows (1.1°C in the first 9 h), and the response was more severe in EL-LPS relative to ML-LPS cows (2.3 vs. 1.3°C increase at 4 h post-LPS; respectively). Respiration rate increased only in EL-LPS cows (47% relative to ML-LPS in the first hour post-LPS). Circulating tumor necrosis factor-α, IL-6, monocyte chemoattractant protein-1, macrophage inflammatory protein (MIP)-1α, MIP-1β, and IFN-γ-inducible protein-10 increased within the first 6 h after LPS and these changes were exacerbated in EL-LPS relative to ML-LPS cows (6.3-fold, 4.8-fold, 57%, 93%, 10%, and 61%, respectively). All cows administered LPS had decreased circulating iCa relative to PF cows (34% at the 6 h nadir), but the hypocalcemia was more severe in EL-LPS than ML-LPS cows (14% at 6 h nadir). In response to LPS, neutrophils decreased regardless of LS, then increased into neutrophilia by 24 h in all LPS relative to PF cows (2-fold); however, the neutrophilic phase was augmented in EL- compared with ML-LPS cows (63% from 24 to 72 h). Lymphocytes and monocytes rapidly decreased then gradually returned to baseline in LPS cows regardless of LS; however, monocytes were increased (57%) at 72 h in EL-LPS relative to ML-LPS cows. Platelets were reduced (46%) in LPS relative to PF cows throughout the 3-d following LPS, and from 24 to 48 h, platelets were further decreased (41%) in EL-LPS compared with ML-LPS. During the 3-d following LPS, serum amyloid A (SAA), LPS-binding protein (LBP), and haptoglobin (Hp) increased in LPS compared with PF groups (9-fold, 72%, and 153-fold, respectively), and the LBP and Hp responses were more exaggerated in EL-LPS than ML-LPS cows (85 and 79%, respectively) whereas the SAA response did not differ by LS. Thus, our data indicates that EL immune function does not appear "suppressed," and in fact many aspects of the immune response are seemingly functionally robust.
Collapse
Affiliation(s)
- J Opgenorth
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - E J Mayorga
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - M A Abeyta
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - B M Goetz
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | | | - A D Freestone
- Department of Animal Science, Iowa State University, Ames, IA 50011
| | - J L McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011
| | - L H Baumgard
- Department of Animal Science, Iowa State University, Ames, IA 50011.
| |
Collapse
|
136
|
Liu Y, Wenren M, Cheng W, Zhou X, Xu D, Chi C, Lü Z, Liu H. Identification, functional characterization and immune response profiles of interleukin-10 in Nibea albiflora. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109654. [PMID: 38810711 DOI: 10.1016/j.fsi.2024.109654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024]
Abstract
Interleukin-10 (IL-10) is an immunosuppressive cytokine, which plays a vital role in regulating inflammation for inhibiting the generation and function of pro-inflammatory cytokines in vivo or in vitro. In the present study, the full length cDNA of IL-10 was characterized from Nibea albiflora (named as NaIL-10) of 1238 base pairs (bp), containing a 5'-UTR (untranslated region) of 350 bp, a 3'-UTR of 333 bp and an open reading frame (ORF) of 555 bp (Fig. 1A) to encode 184 amino acid residues with a signal peptide at the N-terminus. The sequence analysis showed that NaIL-10 possessed the typical IL-10 family symbolic motif and conversed cysteine residues, similar to its teleost orthologues. Real-time PCR indicated that NaIL-10 had wide distribution in different healthy tissues, with a relatively high expression in immune-related tissues (head kidney, spleen, kidney, liver and gill). Significantly, up-regulations of NaIL-10 after infection against Vibrio parahaemolyticus, Vibrio alginolyticus and Poly I:C were also observed. Subcellular localization manifested that NaIL-10 mainly distributed in the cytoplasm unevenly and aggregately, and there was also a small amount on the cell membrane, indicating that NaIL-10 was secreted to the extracellular space as the known IL-10 homologous molecules. It could co-locate with IL-10 Rα on the membrane of HEK293T cells for their potential interaction, and GST pull-down and Co-IP studies certified the specific and direct interaction between NaIL-10 and NaIL-10 Rα, confirming that an IL-10 ligand-receptor system existed in N.albiflora. The expression of pro-inflammatory cytokines, including TNF-α, IL-6, IL-1β, were dramatically inhibited in LPS-stimulated RAW264.7 macrophages pre-incubated with recombinant NaIL-10 protein, demonstrating its anti-inflammatory roles. Taken together, the results demonstrated the existence of IL-10 ligand-receptor system in N.albiflora for the first time, and indicated the suppressive function of NaIL-10 on pro-inflammatory cytokine expression in inflammatory response, which would be conducive to better comprehending the role of IL-10 in the immunomodulatory mechanisms of teleost.
Collapse
Affiliation(s)
- Yue Liu
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Mingming Wenren
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Wei Cheng
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Xu Zhou
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Dongdong Xu
- Zhejiang Marine Fisheries Research Institute, Key Lab of Mariculture and Enhancement of Zhejiang province, Zhoushan, 316100, China
| | - Changfeng Chi
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Zhenming Lü
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China
| | - Huihui Liu
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, China.
| |
Collapse
|
137
|
Cebesoy EI, Altaca M, Kocak-Oztug NA, Bingül I, Cifcibasi E. Associations between interleukin-10, -12, and - 18 and periodontal health and disease: a cross-sectional study. Clin Oral Investig 2024; 28:458. [PMID: 39080003 PMCID: PMC11289060 DOI: 10.1007/s00784-024-05843-8] [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: 01/20/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024]
Abstract
OBJECTIVE We assessed the levels of Interleukin-10 (IL-10), Interleukin-12 (IL-12), and Interleukin-18 (IL-18) in the gingival crevicular fluid (GCF) of subjects with advanced periodontitis (SIII-SIV) compared to healthy controls and evaluated their correlations with clinical measurements. METHODS This cross-sectional study involved subjects (n = 60) diagnosed with stage III grade B-C (n = 13) to stage IV grade C (n = 17) periodontitis, and periodontally healthy controls (n = 30). Clinical periodontal measurements involved full-mouth. The concentrations of IL-10, IL-12, and IL-18 were determined using enzyme-linked immunosorbent assay (ELISA). RESULTS There were no significant differences in IL-12 level and IL-18/IL-10 ratio between the healthy and periodontitis groups (p = 0.413, p = 0.636, respectively). The IL-10 and IL-18 levels were significantly higher in the periodontitis group than in controls (p < 0.001, p < 0.001, respectively). Significant associations were observed between the periodontitis and IL-10 and IL-18 levels (OR = 1.46, %95 CI 1.19-1.795; OR = 1.13, %95 CI 1.059-1.207, respectively) (p < 0.001, p < 0.001, respectively). CONCLUSIONS There was a correlation between pocket depth and the presence of IL-18 and a strong association between periodontitis and a high level of IL-18. However, there were no direct correlations among the three biomarkers and IL-18/IL-10 ratio, indicating that their roles in periodontal health are complex and multidimensional. CLINICAL RELEVANCE Understanding the cytokine dynamics in GCF provides valuable insights into their potential clinical implications for periodontal disease diagnosis, risk assessment, and tailored therapeutic interventions.
Collapse
Affiliation(s)
- Elif Ilke Cebesoy
- Faculty of Dentistry, Department of Periodontology, Istanbul University, Istanbul, 34116, Turkey
- Institute of Graduate Studies in Health Sciences, Department of Periodontology, Istanbul University, Istanbul, 34126, Turkey
| | - Müge Altaca
- Faculty of Dentistry, Department of Periodontology, Istanbul University, Istanbul, 34116, Turkey
- Institute of Graduate Studies in Health Sciences, Department of Periodontology, Istanbul University, Istanbul, 34126, Turkey
| | - Necla Asli Kocak-Oztug
- Faculty of Dentistry, Department of Periodontology, Istanbul University, Istanbul, 34116, Turkey
- School of Dentistry, Faculty of Health and Behavioural Sciences, The University of Queensland, Brisbane, QLD, 4006, Australia
| | - Ilknur Bingül
- Department of Medical Biochemistry, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Emine Cifcibasi
- Faculty of Dentistry, Department of Periodontology, Istanbul University, Istanbul, 34116, Turkey.
| |
Collapse
|
138
|
Möttönen M, Teräsjärvi J, Rahikkala H, Kvist S, Mertsola J, He Q. Association of IL-17A and IL-10 Polymorphisms with Juvenile Idiopathic Arthritis in Finnish Children. Int J Mol Sci 2024; 25:8323. [PMID: 39125893 PMCID: PMC11311899 DOI: 10.3390/ijms25158323] [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: 05/16/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
To analyze the role of interleukin IL-17A and IL-10 polymorphisms in susceptibility to juvenile idiopathic arthritis (JIA), 98 Finnish children and adolescents with JIA were studied. Data from the 1000 Genomes Project, consisting of 99 healthy Finns, served as the controls. The patients were analyzed for four IL-17A and three IL-10 gene-promoter polymorphisms, and the serum IL-17A, IL-17F, IL-10, and IL-6 levels were determined. The IL-17A rs8193036 variant genotypes (CT/CC) were more common among the patients than controls, especially in those with polyarthritis (OR 1.93, 95% CI 1.11-3.36; p = 0.020). IL-17A rs2275913 minor allele A was more common in patients (OR 1.45, 95% Cl 1.08-1.94; p = 0.014) and especially among patients with oligoarthritis and polyarthritis than the controls (OR 1.61, 95%CI 1.06-2.43; p = 0.024). Carriers of the IL-17A rs4711998 variant genotype (AG/AA) had higher serum IL-17A levels than those with genotype GG. However, carriers of the variant genotypes of IL-17A rs9395767 and rs4711998 appeared to have higher IL-17F levels than those carrying wildtype. IL-10 rs1800896 variant genotypes (TC/CC) were more abundant in patients than in the controls (OR 1.97, 95%CI 1.06-3.70; p = 0.042). Carriers of the IL-10 rs1800896 variant genotypes had lower serum levels of IL-17F than those with wildtype. These data provide preliminary evidence of the roles of IL-17 and IL-10 in the pathogenesis of JIA and its subtypes in the Finnish population. However, the results should be interpreted with caution, as the number of subjects included in this study was limited.
Collapse
Affiliation(s)
- Milja Möttönen
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, 20520 Turku, Finland; (M.M.); (H.R.); (J.M.)
| | - Johanna Teräsjärvi
- Institute of Biomedicine, Research Centre for Infections and Immunity, University of Turku, 20520 Turku, Finland; (J.T.); (S.K.)
| | - Heidi Rahikkala
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, 20520 Turku, Finland; (M.M.); (H.R.); (J.M.)
- Research Unit of Clinical Medicine, University of Oulu, 90014 Oulu, Finland
| | - Sonja Kvist
- Institute of Biomedicine, Research Centre for Infections and Immunity, University of Turku, 20520 Turku, Finland; (J.T.); (S.K.)
| | - Jussi Mertsola
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, 20520 Turku, Finland; (M.M.); (H.R.); (J.M.)
| | - Qiushui He
- Institute of Biomedicine, Research Centre for Infections and Immunity, University of Turku, 20520 Turku, Finland; (J.T.); (S.K.)
- InFLAMES Research Flagship Centre, University of Turku, 20520 Turku, Finland
| |
Collapse
|
139
|
Finnegan D, Connolly C, Mechoud MA, FitzGerald JA, Beresford T, Mathur H, Brennan L, Cotter PD, Loscher CE. Novel Dairy Fermentates Have Differential Effects on Key Immune Responses Associated with Viral Immunity and Inflammation in Dendritic Cells. Foods 2024; 13:2392. [PMID: 39123583 PMCID: PMC11311654 DOI: 10.3390/foods13152392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Fermented foods and ingredients, including furmenties derived from lactic acid bacteria (LAB) in dairy products, can modulate the immune system. Here, we describe the use of reconstituted skimmed milk powder to generate novel fermentates from Lactobacillus helveticus strains SC232, SC234, SC212, and SC210, and from Lacticaseibacillus casei strains SC209 and SC229, and demonstrate, using in vitro assays, that these fermentates can differentially modulate cytokine secretion via bone-marrow-derived dendritic cells (BMDCs) when activated with either the viral ligand loxoribine or an inflammatory stimulus, lipopolysaccharide. Specifically, we demonstrate that SC232 and SC234 increase cytokines IL-6, TNF-α, IL-12p40, IL-23, IL-27, and IL-10 and decrease IL-1β in primary bone-marrow-derived dendritic cells (BMDCs) stimulated with a viral ligand. In contrast, exposure of these cells to SC212 and SC210 resulted in increased IL-10, IL-1β, IL-23, and decreased IL-12p40 following activation of the cells with the inflammatory stimulus LPS. Interestingly, SC209 and SC229 had little or no effect on cytokine secretion by BMDCs. Overall, our data demonstrate that these novel fermentates have specific effects and can differentially enhance key immune mechanisms that are critical to viral immune responses, or can suppress responses involved in chronic inflammatory conditions, such as ulcerative colitis (UC), and Crohn's disease (CD).
Collapse
Affiliation(s)
- Dearbhla Finnegan
- School of Biotechnology, Dublin City University, D09 DX63 Dublin, Ireland;
- Food for Health Ireland, Science Centre South (S2.79), University College Dublin, Dublin 4, Ireland; (C.C.); (M.A.M.); (J.A.F.); (T.B.); (H.M.); (L.B.); (P.D.C.)
| | - Claire Connolly
- Food for Health Ireland, Science Centre South (S2.79), University College Dublin, Dublin 4, Ireland; (C.C.); (M.A.M.); (J.A.F.); (T.B.); (H.M.); (L.B.); (P.D.C.)
- UCD School of Agriculture and Food Science, University College Dublin, D04V1W8 Dublin, Ireland
| | - Monica A. Mechoud
- Food for Health Ireland, Science Centre South (S2.79), University College Dublin, Dublin 4, Ireland; (C.C.); (M.A.M.); (J.A.F.); (T.B.); (H.M.); (L.B.); (P.D.C.)
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Co. Cork, Ireland
| | - Jamie A. FitzGerald
- Food for Health Ireland, Science Centre South (S2.79), University College Dublin, Dublin 4, Ireland; (C.C.); (M.A.M.); (J.A.F.); (T.B.); (H.M.); (L.B.); (P.D.C.)
- UCD School of Agriculture and Food Science, University College Dublin, D04V1W8 Dublin, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Co. Cork, Ireland
| | - Tom Beresford
- Food for Health Ireland, Science Centre South (S2.79), University College Dublin, Dublin 4, Ireland; (C.C.); (M.A.M.); (J.A.F.); (T.B.); (H.M.); (L.B.); (P.D.C.)
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Co. Cork, Ireland
| | - Harsh Mathur
- Food for Health Ireland, Science Centre South (S2.79), University College Dublin, Dublin 4, Ireland; (C.C.); (M.A.M.); (J.A.F.); (T.B.); (H.M.); (L.B.); (P.D.C.)
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Co. Cork, Ireland
| | - Lorraine Brennan
- Food for Health Ireland, Science Centre South (S2.79), University College Dublin, Dublin 4, Ireland; (C.C.); (M.A.M.); (J.A.F.); (T.B.); (H.M.); (L.B.); (P.D.C.)
- UCD School of Agriculture and Food Science, University College Dublin, D04V1W8 Dublin, Ireland
| | - Paul D. Cotter
- Food for Health Ireland, Science Centre South (S2.79), University College Dublin, Dublin 4, Ireland; (C.C.); (M.A.M.); (J.A.F.); (T.B.); (H.M.); (L.B.); (P.D.C.)
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Co. Cork, Ireland
- APC Microbiome Ireland, Biosciences Institute, Biosciences Research Institute, University College Cork, T12 R229 Cork, Ireland
- VistaMilk, P61 C996 Co. Cork, Ireland
| | - Christine E. Loscher
- School of Biotechnology, Dublin City University, D09 DX63 Dublin, Ireland;
- Food for Health Ireland, Science Centre South (S2.79), University College Dublin, Dublin 4, Ireland; (C.C.); (M.A.M.); (J.A.F.); (T.B.); (H.M.); (L.B.); (P.D.C.)
| |
Collapse
|
140
|
Pratticò F, Garajová I. Focus on Pancreatic Cancer Microenvironment. Curr Oncol 2024; 31:4241-4260. [PMID: 39195299 PMCID: PMC11352508 DOI: 10.3390/curroncol31080316] [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: 06/16/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Pancreatic ductal adenocarcinoma remains one of the most lethal solid tumors due to its local aggressiveness and metastatic potential, with a 5-year survival rate of only 13%. A robust connection between pancreatic cancer microenvironment and tumor progression exists, as well as resistance to current anticancer treatments. Pancreatic cancer has a complex tumor microenvironment, characterized by an intricate crosstalk between cancer cells, cancer-associated fibroblasts and immune cells. The complex composition of the tumor microenvironment is also reflected in the diversity of its acellular components, such as the extracellular matrix, cytokines, growth factors and secreted ligands involved in signaling pathways. Desmoplasia, the hallmark of the pancreatic cancer microenvironment, contributes by creating a dense and hypoxic environment that promotes further tumorigenesis, provides innate systemic resistance and suppresses anti-tumor immune invasion. We discuss the complex crosstalk among tumor microenvironment components and explore therapeutic strategies and opportunities in pancreatic cancer research. Better understanding of the tumor microenvironment and its influence on pancreatic cancer progression could lead to potential novel therapeutic options, such as integration of immunotherapy and cytokine-targeted treatments.
Collapse
Affiliation(s)
| | - Ingrid Garajová
- Medical Oncology Unit, University Hospital of Parma, 43100 Parma, Italy;
| |
Collapse
|
141
|
González A, Fullaondo A, Odriozola A. Microbiota-associated mechanisms in colorectal cancer. ADVANCES IN GENETICS 2024; 112:123-205. [PMID: 39396836 DOI: 10.1016/bs.adgen.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide, ranking third in terms of incidence and second as a cause of cancer-related death. There is growing scientific evidence that the gut microbiota plays a key role in the initiation and development of CRC. Specific bacterial species and complex microbial communities contribute directly to CRC pathogenesis by promoting the neoplastic transformation of intestinal epithelial cells or indirectly through their interaction with the host immune system. As a result, a protumoural and immunosuppressive environment is created conducive to CRC development. On the other hand, certain bacteria in the gut microbiota contribute to protection against CRC. In this chapter, we analysed the relationship of the gut microbiota to CRC and the associations identified with specific bacteria. Microbiota plays a key role in CRC through various mechanisms, such as increased intestinal permeability, inflammation and immune system dysregulation, biofilm formation, genotoxin production, virulence factors and oxidative stress. Exploring the interaction between gut microbiota and tumourigenesis is essential for developing innovative therapeutic approaches in the fight against CRC.
Collapse
Affiliation(s)
- Adriana González
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain.
| | - Asier Fullaondo
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| | - Adrian Odriozola
- Hologenomics Research Group, Department of Genetics, Physical Anthropology, and Animal Physiology, University of the Basque Country, Spain
| |
Collapse
|
142
|
Yi M, Li T, Niu M, Zhang H, Wu Y, Wu K, Dai Z. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther 2024; 9:176. [PMID: 39034318 PMCID: PMC11275440 DOI: 10.1038/s41392-024-01868-3] [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] [Revised: 04/30/2024] [Accepted: 05/11/2024] [Indexed: 07/23/2024] Open
Abstract
Cytokines are critical in regulating immune responses and cellular behavior, playing dual roles in both normal physiology and the pathology of diseases such as cancer. These molecules, including interleukins, interferons, tumor necrosis factors, chemokines, and growth factors like TGF-β, VEGF, and EGF, can promote or inhibit tumor growth, influence the tumor microenvironment, and impact the efficacy of cancer treatments. Recent advances in targeting these pathways have shown promising therapeutic potential, offering new strategies to modulate the immune system, inhibit tumor progression, and overcome resistance to conventional therapies. In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer. By exploring the roles of these molecules in tumor biology and the immune response, we highlighted the development of novel therapeutic agents aimed at modulating these pathways to combat cancer. The review elaborated on the dual nature of cytokines as both promoters and suppressors of tumorigenesis, depending on the context, and discussed the challenges and opportunities this presents for therapeutic intervention. We also examined the latest advancements in targeted therapies, including monoclonal antibodies, bispecific antibodies, receptor inhibitors, fusion proteins, engineered cytokine variants, and their impact on tumor growth, metastasis, and the tumor microenvironment. Additionally, we evaluated the potential of combining these targeted therapies with other treatment modalities to overcome resistance and improve patient outcomes. Besides, we also focused on the ongoing research and clinical trials that are pivotal in advancing our understanding and application of cytokine- and chemokine-targeted therapies for cancer patients.
Collapse
Affiliation(s)
- Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Haoxiang Zhang
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|
143
|
Liu Y, Yang J, Guo Z, Li Q, Zhang L, Zhao L, Zhou X. Immunomodulatory Effect of Cordyceps militaris Polysaccharide on RAW 264.7 Macrophages by Regulating MAPK Signaling Pathways. Molecules 2024; 29:3408. [PMID: 39064986 PMCID: PMC11279930 DOI: 10.3390/molecules29143408] [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: 05/22/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Polysaccharide is one of the principal bioactive components found in medicinal mushrooms and has been proven to enhance host immunity. However, the possible mechanism of immunomodulatory activity of Cordyceps militaris polysaccharide is not fully understood. Hot water extraction and alcohol precipitation, DEAE-Sephadex A-25 chromatography, and Sephadex G-100 chromatography were used to isolate polysaccharide from C. militaris. A high-molecular-weight polysaccharide isolated from C. militaris was designated as HCMP, which had an Mw of 6.18 × 105 Da and was composed of arabinose, galactose, glucose, mannose, and xylose in a mole ratio of 2.00:8.01:72.54:15.98:1.02. The polysaccharide content of HCMP was 91.2% ± 0.16. The test in vitro showed that HCMP activated mouse macrophage RAW 264.7 cells by enhancing phagocytosis and NO production, and by regulating mRNA expressions of inflammation-related molecules in RAW 264.7 cells. Western blotting revealed that HCMP induced the phosphorylation of mitogen-activated protein kinases (MAPKs). Moreover, using inhibitors of MAPKs decreased the mRNA levels of inflammation-related molecules induced by HCMP. These data evidenced that the immunomodulatory effect of HCMP on RAW 264.7 macrophages was mediated via the MAPK signaling pathway. These findings suggested that HCMP could be developed as a potent immunomodulatory agent for use in functional foods and dietary supplements.
Collapse
Affiliation(s)
- Yan Liu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.L.); (J.Y.); (Z.G.)
- Engineering Research Center of Therapeutic Antibody (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiayi Yang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.L.); (J.Y.); (Z.G.)
- Engineering Research Center of Therapeutic Antibody (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhijian Guo
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.L.); (J.Y.); (Z.G.)
- Engineering Research Center of Therapeutic Antibody (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qizhang Li
- Innovative Drug R&D Center, College of Life Sciences, Huaibei Normal University, Huaibei 235000, China
| | - Lida Zhang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.L.); (J.Y.); (Z.G.)
- Engineering Research Center of Therapeutic Antibody (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lingxia Zhao
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.L.); (J.Y.); (Z.G.)
- Engineering Research Center of Therapeutic Antibody (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xuanwei Zhou
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.L.); (J.Y.); (Z.G.)
- Engineering Research Center of Therapeutic Antibody (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
144
|
Kraxner A, Braun F, Cheng WY, Yang THO, Pipaliya S, Canamero M, Andersson E, Harring SV, Dziadek S, Bröske AME, Ceppi M, Tanos T, Teichgräber V, Charo J. Investigating the complex interplay between fibroblast activation protein α-positive cancer associated fibroblasts and the tumor microenvironment in the context of cancer immunotherapy. Front Immunol 2024; 15:1352632. [PMID: 39035007 PMCID: PMC11258004 DOI: 10.3389/fimmu.2024.1352632] [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: 12/08/2023] [Accepted: 06/19/2024] [Indexed: 07/23/2024] Open
Abstract
Introduction This study investigates the role of Fibroblast Activation Protein (FAP)-positive cancer-associated fibroblasts (FAP+CAF) in shaping the tumor immune microenvironment, focusing on its association with immune cell functionality and cytokine expression patterns. Methods Utilizing immunohistochemistry, we observed elevated FAP+CAF density in metastatic versus primary renal cell carcinoma (RCC) tumors, with higher FAP+CAF correlating with increased T cell infiltration in RCC, a unique phenomenon illustrating the complex interplay between tumor progression, FAP+CAF density, and immune response. Results Analysis of immune cell subsets in FAP+CAF-rich stromal areas further revealed significant correlations between FAP+ stroma and various T cell types, particularly in RCC and non-small cell lung cancer (NSCLC). This was complemented by transcriptomic analyses, expanding the range of stromal and immune cell subsets interrogated, as well as to additional tumor types. This enabled evaluating the association of these subsets with tumor infiltration, tumor vascularization and other components of the tumor microenvironment. Our comprehensive study also encompassed cytokine, angiogenesis, and inflammation gene signatures across different cancer types, revealing heterogeneous cellular composition, cytokine expressions and angiogenic profiles. Through cytokine pathway profiling, we explored the relationship between FAP+CAF density and immune cell states, uncovering potential immunosuppressive circuits that limit anti-tumor activity in tumor-resident immune cells. Conclusions These findings underscore the complexity of tumor biology and the necessity for personalized therapeutic and patient enrichment approaches. The insights gathered from FAP+CAF prevalence, immune infiltration, and gene signatures provide valuable perspectives on tumor microenvironments, aiding in future research and clinical strategy development.
Collapse
Affiliation(s)
- Anton Kraxner
- Roche Pharma Research and Early Development, Oncology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Franziska Braun
- Roche Pharma Research and Early Development, Data and Analytics, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Wei-Yi Cheng
- Roche Pharma Research and Early Development, Data and Analytics, Roche Translational & Clinical Research Center, F. Hoffmann-La Roche Ltd, Little Falls, NJ, United States
| | - Tai-Hsien Ou Yang
- Roche Pharma Research and Early Development, Data and Analytics, Roche Translational & Clinical Research Center, F. Hoffmann-La Roche Ltd, Little Falls, NJ, United States
| | - Shweta Pipaliya
- Roche Pharma Research and Early Development, Data and Analytics, Roche Innovation Center Zurich, Roche Glycart AG, Schlieren, Switzerland
| | - Marta Canamero
- Roche Pharma Research and Early Development, Oncology, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Emilia Andersson
- Roche Pharma Research and Early Development, Oncology, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Suzana Vega Harring
- Roche Pharma Research and Early Development, Oncology, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Sebastian Dziadek
- Roche Pharma Research and Early Development, Oncology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Ann-Marie E. Bröske
- Roche Pharma Research and Early Development, Oncology, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Maurizio Ceppi
- Roche Pharma Research and Early Development, Oncology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Tamara Tanos
- Roche Pharma Research and Early Development, Oncology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Volker Teichgräber
- Roche Pharma Research and Early Development, Oncology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Jehad Charo
- Roche Pharma Research and Early Development, Oncology, Roche Innovation Center Zurich, Roche Glycart AG, Schlieren, Switzerland
| |
Collapse
|
145
|
Sun Y, Wu D, Yang X, Tang B, Xia C, Luo C, Gong Q, Lui S, Hu N. The associations of peripheral interleukin alterations and hippocampal subfield volume deficits in schizophrenia. Cereb Cortex 2024; 34:bhae308. [PMID: 39077921 DOI: 10.1093/cercor/bhae308] [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: 05/08/2024] [Revised: 07/03/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
The hippocampus is one of the brain regions most vulnerable to inflammatory insults, and the relationships between peripheral inflammation and hippocampal subfields in patients with schizophrenia remain unclear. In this study, forty-six stably medicated patients with schizophrenia and 48 demographically matched healthy controls (HCs) were recruited. The serum levels of IL - 1β, IL-6, IL-10, and IL-12p70 were measured, and 3D high-resolution T1-weighted magnetic resonance imaging was performed. The IL levels and hippocampal subfield volumes were both compared between patients and HCs. The associations of altered IL levels with hippocampal subfield volumes were assessed in patients. Patients with schizophrenia demonstrated higher serum levels of IL-6 and IL-10 but lower levels of IL-12p70 than HCs. In patients, the levels of IL-6 were positively correlated with the volumes of the left granule cell layer of the dentate gyrus (GCL) and cornu Ammonis (CA) 4, while the levels of IL-10 were negatively correlated with the volumes of those subfields. IL-6 and IL-10 might have antagonistic roles in atrophy of the left GCL and CA4. This suggests a complexity of peripheral cytokine dysregulation and the potential for its selective effects on hippocampal substructures, which might be related to the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Yuan Sun
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Dongsheng Wu
- Department of Radiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 18, Section 3, South Renmin Road, Chengdu 610041, China
| | - Xiyue Yang
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Biqiu Tang
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Chao Xia
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Chunyan Luo
- Department of Radiology, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Su Lui
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| | - Na Hu
- Department of Radiology, and Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
- Department of Radiology, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 610041, China
| |
Collapse
|
146
|
Payen SH, Adhikari K, Petereit J, Uppal T, Rossetto CC, Verma SC. SARS-CoV-2 superinfection in CD14 + monocytes with latent human cytomegalovirus (HCMV) promotes inflammatory cascade. Virus Res 2024; 345:199375. [PMID: 38642618 PMCID: PMC11061749 DOI: 10.1016/j.virusres.2024.199375] [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: 02/16/2024] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiologic agent of coronavirus disease 2019 (COVID-19), has posed significant challenges to global health. While much attention has been directed towards understanding the primary mechanisms of SARS-CoV-2 infection, emerging evidence suggests co-infections or superinfections with other viruses may contribute to increased morbidity and mortality, particularly in severe cases of COVID-19. Among viruses that have been reported in patients with SARS-CoV-2, seropositivity for Human cytomegalovirus (HCMV) is associated with increased COVID-19 risk and hospitalization. HCMV is a ubiquitous beta-herpesvirus with a seroprevalence of 60-90 % worldwide and one of the leading causes of mortality in immunocompromised individuals. The primary sites of latency for HCMV include CD14+ monocytes and CD34+ hematopoietic cells. In this study, we sought to investigate SARS-CoV-2 infection of CD14+ monocytes latently infected with HCMV. We demonstrate that CD14+ cells are susceptible and permissive to SARS-CoV-2 infection and detect subgenomic transcripts indicative of replication. To further investigate the molecular changes triggered by SARS-CoV-2 infection in HCMV-latent CD14+ monocytes, we conducted RNA sequencing coupled with bioinformatic differential gene analysis. The results revealed significant differences in cytokine-cytokine receptor interactions and inflammatory pathways in cells superinfected with replication-competent SARS-CoV-2 compared to the heat-inactivated and mock controls. Notably, there was a significant upregulation in transcripts associated with pro-inflammatory response factors and a decrease in anti-inflammatory factors. Taken together, these findings provide a basis for the heightened inflammatory response, offering potential avenues for targeted therapeutic interventions among HCMV-infected severe cases of COVID-19. SUMMARY: COVID-19 patients infected with secondary viruses have been associated with a higher prevalence of severe symptoms. Individuals seropositive for human cytomegalovirus (HCMV) infection are at an increased risk for severe COVID-19 disease and hospitalization. HCMV reactivation has been reported in severe COVID-19 cases with respiratory failure and could be the result of co-infection with SARS-CoV-2 and HCMV. In a cell culture model of superinfection, HCMV has previously been shown to increase infection of SARS-CoV-2 of epithelial cells by upregulating the human angiotensin-converting enzyme-2 (ACE2) receptor. In this study, we utilize CD14+ monocytes, a major cell type that harbors latent HCMV, to investigate co-infection of SARS-CoV-2 and HCMV. This study is a first step toward understanding the mechanism that may facilitate increased COVID-19 disease severity in patients infected with SARS-CoV-2 and HCMV.
Collapse
Affiliation(s)
- Shannon Harger Payen
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States
| | - Kabita Adhikari
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States
| | - Juli Petereit
- Nevada Bioinformatics Center (RRID:SCR_017802), University of Nevada, Reno, NV 89557, United States
| | - Timsy Uppal
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States
| | - Cyprian C Rossetto
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States
| | - Subhash C Verma
- Reno School of Medicine, Department of Microbiology & Immunology/MS 320, University of Nevada, Reno, NV 89557, United States.
| |
Collapse
|
147
|
Sheth RA, Wehrenberg-Klee E, Patel SP, Brock KK, Fotiadis N, de Baère T. Intratumoral Injection of Immunotherapeutics: State of the Art and Future Directions. Radiology 2024; 312:e232654. [PMID: 39078294 PMCID: PMC11294769 DOI: 10.1148/radiol.232654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 02/17/2024] [Accepted: 02/28/2024] [Indexed: 07/31/2024]
Abstract
Systemic immunotherapies have led to tremendous progress across the cancer landscape. However, several challenges exist, potentially limiting their efficacy in the treatment of solid tumors. Direct intratumoral injection can increase the therapeutic index of immunotherapies while overcoming many of the barriers associated with systemic administration, including limited bioavailability to tumors and potential systemic safety concerns. However, challenges remain, including the lack of standardized approaches for administration, issues relating to effective drug delivery, logistical hurdles, and safety concerns specific to this mode of administration. This article reviews the biologic rationale for the localized injection of immunotherapeutic agents into tumors. It also addresses the existing limitations and practical considerations for safe and effective implementation and provide recommendations for optimizing logistics and treatment workflows. It also highlights the critical role that radiologists, interventional radiologists, and medical physicists play in intratumoral immunotherapy with respect to target selection, image-guided administration, and response assessment.
Collapse
Affiliation(s)
- Rahul A. Sheth
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Eric Wehrenberg-Klee
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Sapna P. Patel
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Kristy K. Brock
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Nicos Fotiadis
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Thierry de Baère
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| |
Collapse
|
148
|
Andres-Martin F, James C, Catalfamo M. IL-27 expression regulation and its effects on adaptive immunity against viruses. Front Immunol 2024; 15:1395921. [PMID: 38966644 PMCID: PMC11222398 DOI: 10.3389/fimmu.2024.1395921] [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: 03/04/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024] Open
Abstract
IL-27, a member of the IL-6/IL-12 cytokine superfamily, is primarily secreted by antigen presenting cells, specifically by dendric cells, macrophages and B cells. IL-27 has antiviral activities and modulates both innate and adaptive immune responses against viruses. The role of IL-27 in the setting of viral infections is not well defined and both pro-inflammatory and anti-inflammatory functions have been described. Here, we discuss the latest advancements in the role of IL-27 in several viral infection models of human disease. We highlight important aspects of IL-27 expression regulation, the critical cell sources at different stages of the infection and their impact in cell mediated immunity. Lastly, we discuss the need to better define the antiviral and modulatory (pro-inflammatory vs anti-inflammatory) properties of IL-27 in the context of human chronic viral infections.
Collapse
Affiliation(s)
| | | | - Marta Catalfamo
- Department of Microbiology Immunology, Georgetown University School of Medicine, Washington, DC, United States
| |
Collapse
|
149
|
Obare LM, Simmons J, Oakes J, Zhang X, Nochowicz C, Priest S, Bailin SS, Warren CM, Mashayekhi M, Beasley HK, Shao J, Meenderink LM, Sheng Q, Stolze J, Gangula R, Absi T, Su YR, Neikirk K, Chopra A, Gabriel CL, Temu T, Pakala S, Wilfong EM, Gianella S, Phillips EJ, Harrison DG, Hinton A, Kalams SA, Kirabo A, Mallal SA, Koethe JR, Wanjalla CN. CD3 + T-cell: CD14 +monocyte complexes are dynamic and increased with HIV and glucose intolerance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.24.538020. [PMID: 37162990 PMCID: PMC10168203 DOI: 10.1101/2023.04.24.538020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
An increased risk of cardiometabolic disease accompanies persistent systemic inflammation. Yet, the innate and adaptive immune system features in persons who develop these conditions remain poorly defined. Doublets, or cell-cell complexes, are routinely eliminated from flow cytometric and other immune phenotyping analyses, which limits our understanding of their relationship to disease states. Using well-characterized clinical cohorts, including participants with controlled HIV as a model for chronic inflammation and increased immune cell interactions, we show that circulating CD14+ monocytes complexed to CD3+ T cells are dynamic, biologically relevant, and increased in individuals with diabetes after adjusting for confounding factors. The complexes form functional immune synapses with increased expression of proinflammatory cytokines and greater glucose utilization. Furthermore, in persons with HIV, the CD3+T-cell: CD14+monocyte complexes had more HIV copies compared to matched CD14+ monocytes or CD4+ T cells alone. Our results demonstrate that circulating CD3+T-cell:CD14+monocyte pairs represent dynamic cellular interactions that may contribute to inflammation and cardiometabolic disease pathogenesis and may originate or be maintained, in part, by chronic viral infections. These findings provide a foundation for future studies investigating mechanisms linking T cellmonocyte cell-cell complexes to developing immune-mediated diseases, including HIV and diabetes.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua Simmons
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jared Oakes
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Xiuqi Zhang
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cindy Nochowicz
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephen Priest
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Samuel S. Bailin
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Mona Mashayekhi
- Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, USA
| | - Leslie M. Meenderink
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - Joey Stolze
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - Rama Gangula
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tarek Absi
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yan Ru Su
- Department of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - Curtis L. Gabriel
- Division of Gastroenterology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tecla Temu
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Suman Pakala
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Erin M. Wilfong
- Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara Gianella
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elizabeth J. Phillips
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David G. Harrison
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Spyros A. Kalams
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Annet Kirabo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - John R. Koethe
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
150
|
Iglesias M, Bibicheff D, Komin A, Chicco M, Guinn S, Foley B, Raimondi G. T cell responsiveness to IL-10 defines the immunomodulatory effect of costimulation blockade via anti-CD154 and impacts transplant survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598652. [PMID: 38915537 PMCID: PMC11195256 DOI: 10.1101/2024.06.12.598652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Costimulation blockade (CoB)-based immunotherapy is a promising alternative to immunosuppression for transplant recipients; however, the current limited understanding of the factors that impact its efficacy restrains its clinical applicability. In this context, pro- and anti-inflammatory cytokines are being recognized as having an impact on T cell activation beyond effector differentiation. This study aims at elucidating the impact of direct IL-10 signaling in T cells on CoB outcomes. We used a full-mismatch skin transplantation model where recipients had a T cell-restricted expression of a dominant negative IL-10 receptor (10R-DN), alongside anti-CD154 as CoB therapy. Unlike wild-type recipients, 10R-DN mice failed to benefit from CoB. This accelerated graft rejection correlated with increased accumulation of T cells producing TNF-α, IFN-γ, and IL-17. In vitro experiments indicated that while lack of IL-10 signaling did not change the ability of anti-CD154 to modulate alloreactive T cell proliferation, the absence of this pathway heightened TH1 effector cell differentiation. Furthermore, deficiency of IL-10 signaling in T cells impaired Treg induction, a hallmark of anti-CD154 therapy. Overall, these findings unveil an important and novel role of IL-10 signaling in T cells that defines the success of CoB therapies and identifies a target pathway for obtaining robust immunoregulation.
Collapse
Affiliation(s)
- Marcos Iglesias
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Darrel Bibicheff
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexander Komin
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maria Chicco
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samantha Guinn
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brendan Foley
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Giorgio Raimondi
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|