Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 18.503
Filter
1.
Prog Orthod ; 25(1): 38, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39370477

ABSTRACT

BACKGROUND: The prevailing paradigm posits orthodontic tooth movement (OTM) as primarily a localized inflammatory process. In this study, we endeavor to elucidate the potential ramifications of mechanical force on systemic immunity, employing a time-dependent approach. MATERIALS AND METHODS: A previously described mouse orthodontic model was used. Ni-Ti. springs were set to move the upper 1st-molar in C57BL/6 mice and the amount of OTM was. measured by µCT. Mice were allocated randomly into four experimental groups, each. corresponding to clinical phases of OTM, relative to force application. Terminal blood. samples were collected and a comprehensive blood count test for 7 cell types as well as. proteome profiling of 111 pivotal cytokines and chemokines were conducted. Two controls. groups were included: one comprised non-treated mice and the other mice with inactivated springs. RESULTS: Serum immuno-profiling unveiled alterations in cellular immunity, manifesting as. changes in percentages of leukocytes, monocytes, macrophages, neutrophils, and. lymphocytes, alongside key signaling factors in comparison to both control groups. The systemic cellular and molecular alterations triggered by OTM mirrored the dynamics previously described in the local immune response. CONCLUSIONS: Although the exact interplay between local and systemic immune responses to orthodontic forces require further elucidation, our findings demonstrate a tangible link between the two. Future investigations should aim to correlate these results with human subjects, and strive to delve deeper into the specific mechanisms by which mechanical force modulates the systemic immune response.


Subject(s)
Mice, Inbred C57BL , Tooth Movement Techniques , Tooth Movement Techniques/methods , Animals , Mice , Cytokines , Immunity, Cellular , X-Ray Microtomography , Macrophages/immunology , Nickel , Neutrophils/immunology , Random Allocation , Chemokines/blood , Titanium
2.
Sci Rep ; 14(1): 23111, 2024 10 04.
Article in English | MEDLINE | ID: mdl-39367102

ABSTRACT

Research into the efficacy of photobiomodulation therapy (PBMT) in reducing inflammation has been ongoing for years, but standards for irradiation methodology still need to be developed. This study aimed to test whether PBMT stimulates in vitro human peripheral blood mononuclear cells (PBMCs) to synthesize pro-inflammatory cytokines, including chemokines. PBMCs were irradiated with laser radiation at two wavelengths simultaneously (λ = 808 nm in continuous emission and λ = 905 nm in pulsed emission). The laser radiation energy was dosed in one dose as a whole (5 J, 15 J, 20 J) or in a fractionated way (5 J + 15 J and 15 J + 5 J) with a frequency of 500, 1,500 and 2,000 Hz. The surface power densities were 177, 214 and 230 mW/cm2, respectively. A pro-inflammatory effect was observed at both the transcript and protein levels for IL-1ß after PBMT at the energy doses 5 J and 20 J (ƒ=500 Hz) and only at the transcript level after application of PBMT at energy doses of 20 J (ƒ= 1,500; ƒ=2,000 Hz) and 5 + 15 J (ƒ=500 Hz). An increase in CCL2 and CCL3 mRNA expression was observed after PBMT at 5 + 15 J (ƒ=1,500 Hz) and 15 + 5 J (ƒ=2,000 Hz) and CCL3 concentration after application of an energy dose of 15 J (frequency of 500 Hz). Even though PBMT can induce mRNA synthesis and stimulate PBMCs to produce selected pro-inflammatory cytokines and chemokines, it is necessary to elucidate the impact of the simultaneous emission of two wavelengths on the inflammatory response mechanisms.


Subject(s)
Inflammation , Leukocytes, Mononuclear , Low-Level Light Therapy , Humans , Leukocytes, Mononuclear/radiation effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , Low-Level Light Therapy/methods , Inflammation/radiotherapy , Cytokines/metabolism , Cells, Cultured , Interleukin-1beta/metabolism , Chemokines/metabolism , Chemokine CCL2/metabolism , Chemokine CCL2/genetics
3.
Blood Press ; 33(1): 2414072, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39387176

ABSTRACT

OBJECTIVE: To study the role of perivascular adipose tissue (PVAT) in the reactivity of rat and human vessels. METHODS: Iliac and mesenteric arteries were obtained from normotensive Sprague-Dawley rats, hypertensive transgenic (mRen2)27 rats overexpressing mouse renin, and (mRen2)27 rats made diabetic with streptozotocin. Human coronary arteries were obtained from donors. Concentration-response curves were constructed to endothelin-1 and acetylcholine with and without PVAT. The contribution of NO and endothelium-dependent hyperpolarization (EDH) were determined making use of the NO synthase inhibitor L-NAME and the EDH inhibitors apamin + TRAM-34. The endothelin type A and type B (ETA, ETB) receptor blockers BQ123 and BQ788, the chemerin inhibitors α-NETA and pravastatin, and the angiotensin receptor blocker losartan were also used. RESULTS: In rat iliac arteries, PVAT diminished endothelin-induced constriction, while the opposite was true in human coronaries. Coronary effects were unaltered by α-NETA, pravastatin, or losartan. ETB receptor-mediated relaxation in iliac arteries occurred only with PVAT, and BQ123 blocked endothelin-1-induced constriction. Diabetes upregulated the anticontractile effects of PVAT. In rat mesenteric arteries, acetylcholine-induced relaxation with PVAT relied on NO, and on NO + EDH without PVAT. Diabetes upregulated the EDH component exclusively with PVAT. CONCLUSION: PVAT modulates ET-1-induced constriction in a vessel type-dependent manner. Its enhancing effects in coronaries involved neither chemerin nor angiotensin II. Its anticontractile effects in rat iliac arteries involved ETB receptor-mediated relaxation. Diabetes upregulated PVAT's anticontractile effects. In mesenteric arteries, PVAT counterbalanced the EDH component of the relaxant effect of acetylcholine. Diabetes reversed this effect by upregulating the EDH component.


What is the context?The role of perivascular adipose tissue (PVAT) in vascular reactivity in pathological conditions is poorly understood.What is the study about?This study investigates the role of PVAT in vascular reactivity in animal and human vessels.What are the results?PVAT has vasoconstrictor and vasorelaxant effects depending on location and tissue. In human coronary arteries, PVAT-mediated vasoconstrictor effects do not involve chemerin or angiotensin II. PVAT's anticontractile effects in rat iliac arteries are mediated through a mechanism involving endothelin type B receptor-dependent relaxation. Moreover, diabetes but not hypertension dysregulates PVAT's anticontractile effects in rat mesenteric vessels.


Subject(s)
Acetylcholine , Adipose Tissue , Angiotensin II , Chemokines , Diabetes Mellitus, Experimental , Endothelin-1 , Mesenteric Arteries , Rats, Sprague-Dawley , Animals , Humans , Endothelin-1/pharmacology , Endothelin-1/metabolism , Rats , Acetylcholine/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/metabolism , Angiotensin II/pharmacology , Adipose Tissue/metabolism , Male , Chemokines/metabolism , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiopathology , Mesenteric Arteries/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Vasoconstriction/drug effects , Mice
4.
Commun Biol ; 7(1): 1304, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39394451

ABSTRACT

SATB1, a key regulator of T cell development, governs lineage-specific transcriptional programs upon T cell activation. The absence of SATB1 has been linked to the initiation and progression of autoimmunity. However, its precise roles in this process remain incompletely understood. Here we show that conditional knockout of Satb1 in CD4+ T cells in mice led to T cell hyperactivation and inflammatory cell infiltration across multiple organs. Transcriptional profiling on activated T cells revealed that the loss of SATB1 led to aberrant upregulation of CC chemokines. Treating Satb1 conditional knockout mice with CC chemokine receptor inhibitor alleviated inflammatory cell infiltration. Intriguingly, SATB1's transcriptional regulation of chemokine genes could not be attributed to its direct binding to chemokine promoters. Instead, SATB1 exerted its regulatory effects by controlling higher-order chromatin organization at a CC chemokine locus. The loss of SATB1 led to the emergence of a new chromatin domain encompassing the Ccl3, Ccl4, Ccl5, Ccl6, and Ccl9 genes and a distal enhancer, resulting in increased contacts between the enhancer and all five chemokine genes, thus inducing their upregulation. Collectively, these results demonstrate that SATB1 protects organs from immune cell infiltration by regulating chemokine expression, providing valuable insights into the development of autoimmunity-related phenotypes.


Subject(s)
Chemokines , Chromatin , Matrix Attachment Region Binding Proteins , Mice, Knockout , Animals , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Mice , Chromatin/metabolism , Chemokines/metabolism , Chemokines/genetics , Gene Expression Regulation , Multigene Family , Mice, Inbred C57BL , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Lymphocyte Activation
5.
BMC Vet Res ; 20(1): 411, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39272157

ABSTRACT

BACKGROUND: Sepsis, defined as a dysregulated inflammatory response to infection inducing organ dysfunction, is a common cause of mortality in both humans and animals. Early detection and treatment is essential for survival, but accurate diagnosis is challenging due to the lack of specific biomarkers for sepsis. This study explored the potential of the keratinocyte-derived chemokine (KC)-like protein in dogs as a biomarker of sepsis in dogs with bacterial uterine infection (pyometra). The aim was to compare KC-like concentrations in dogs with pyometra with or without sepsis and to assess associations between KC-like and clinical variables, including days of hospitalization as an outcome. RESULTS: A mouse KC ELISA was validated and used to determine the concentrations of KC-like in serum from 34 dogs with pyometra and 18 healthy controls. Dogs with pyometra were classified as having sepsis based on two different criteria for systemic inflammatory response syndrome (SIRS), resulting in 74% and 30% sepsis-positive, respectively. The concentration of KC-like protein was higher in pyometra dogs with sepsis than in pyometra dogs without sepsis (p < 0.05) and in healthy controls (p < 0.0001) when using either of the two SIRS criteria. Moreover, KC-like was slightly increased in dogs with pyometra without sepsis compared with healthy controls when using the more stringent SIRS criteria (p < 0.05). Analyses of all dogs showed that KC-like concentrations correlated positively with hospitalization days, C-reactive protein (CRP) concentrations, white blood cells, and percentage of band neutrophils; however, KC-like correlated negatively with hemoglobin and did not correlate with circulating creatinine. CONCLUSIONS: Our results suggest that circulating KC-like protein increases in dogs with sepsis in pyometra and that KC-like is associated with more severe clinical illness. These findings support a potential role of KC-like as a biomarker of sepsis; however, the true identity of KC-like in dogs has yet to be uncovered.


Subject(s)
Biomarkers , Dog Diseases , Pyometra , Sepsis , Animals , Dogs , Pyometra/veterinary , Pyometra/blood , Pyometra/complications , Female , Dog Diseases/blood , Biomarkers/blood , Sepsis/veterinary , Sepsis/blood , Chemokines/blood , Enzyme-Linked Immunosorbent Assay/veterinary , Systemic Inflammatory Response Syndrome/veterinary , Systemic Inflammatory Response Syndrome/blood
6.
Lab Chip ; 24(20): 4892-4906, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39308400

ABSTRACT

The growth of new blood vessels through angiogenesis is a highly coordinated process, which is initiated by chemokine gradients that activate endothelial cells within a perfused parent vessel to sprout into the surrounding 3D tissue matrix. While both biochemical signals from pro-angiogenic factors, as well as mechanical cues originating from luminal fluid flow that exerts shear stress on the vessel wall, have individually been identified as major regulators of endothelial cell sprouting, it remains unclear whether and how both types of cues synergize. To fill this knowledge gap, here, we created a 3D biomimetic model of chemokine gradient-driven angiogenic sprouting, in which a micromolded tube inside a hydrogel matrix is seeded with endothelial cells and connected to a perfusion system to control fluid flow rates and resulting shear forces on the vessel wall. To allow for the formation of chemokine gradients despite the presence of luminal flow, a nanoporous synthetic hydrogel that supports angiogenesis but limits the interstitial flow proved crucial. Using this system, we find that luminal flow and resulting shear stress is a major regulator of the speed and morphogenesis of angiogenic sprouting, whose action is mediated through changes in vascular permeability.


Subject(s)
Human Umbilical Vein Endothelial Cells , Hydrogels , Neovascularization, Physiologic , Humans , Neovascularization, Physiologic/drug effects , Hydrogels/chemistry , Human Umbilical Vein Endothelial Cells/metabolism , Chemokines/metabolism , Nanopores , Models, Biological , Angiogenesis
7.
Sci Signal ; 17(852): eads7460, 2024 09 03.
Article in English | MEDLINE | ID: mdl-39226375

ABSTRACT

Negatively charged lipid bilayers enhance the interaction between a chemokine and an atypical chemokine receptor.


Subject(s)
Lipid Bilayers , Humans , Lipid Bilayers/metabolism , Lipid Bilayers/chemistry , Chemokines/metabolism , Receptors, Chemokine/metabolism
8.
Biomolecules ; 14(9)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39334887

ABSTRACT

Obesity is a global health crisis that is closely interrelated to many chronic diseases, such as cardiovascular disease and diabetes. This review provides an in-depth analysis of specific chemokines involved in the development of obesity, including C-C motif chemokine ligand 2 (CCL2), CCL3, CCL5, CCL7, C-X-C motif chemokine ligand 8 (CXCL8), CXCL9, CXCL10, CXCL14, and XCL1 (lymphotactin). These chemokines exacerbate the symptoms of obesity by either promoting the inflammatory response or by influencing metabolic pathways and recruiting immune cells. Additionally, the research highlights the positive effect of exercise on modulating chemokine expression in the obese state. Notably, it explores the potential effects of both aerobic exercises and combined aerobic and resistance training in lowering levels of inflammatory mediators, reducing insulin resistance, and improving metabolic health. These findings suggest new strategies for obesity intervention through the modulation of chemokine levels by exercise, providing fresh perspectives and directions for the treatment of obesity and future research.


Subject(s)
Chemokines , Exercise , Obesity , Weight Loss , Humans , Obesity/metabolism , Chemokines/metabolism , Animals
9.
Int Rev Cell Mol Biol ; 388: 124-161, 2024.
Article in English | MEDLINE | ID: mdl-39260935

ABSTRACT

Chemokines and their receptors are a family of chemotactic cytokines with important functions in the immune response in both health and disease. Their known physiological roles such as the regulation of leukocyte trafficking and the development of immune organs generated great interest when it was found that they were also related to the control of early and late inflammatory stages in the tumor microenvironment. In fact, in breast cancer, an imbalance in the synthesis of chemokines and/or in the expression of their receptors was attributed to be involved in the regulation of disease progression, including invasion and metastasis. Research in this area is progressing rapidly and the development of new agents based on chemokine and chemokine receptor antagonists are emerging as attractive alternative strategies. This chapter provides a snapshot of the different functions reported for chemokines and their receptors with respect to the potential to regulate breast cancer progression.


Subject(s)
Breast Neoplasms , Disease Progression , Neoplasm Metastasis , Receptors, Chemokine , Humans , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Female , Ligands , Receptors, Chemokine/metabolism , Animals , Chemokines/metabolism
10.
Exp Dermatol ; 33(9): e15173, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39246287

ABSTRACT

In this study, we aimed to examine the relationship between the serum cytokine levels of patients with pemphigus vulgaris (PV) and the Pemphigus Disease Area Index (PDAI), along with the presence of anti-desmoglein (Dsg) 1 antibody, anti-Dsg3 antibody and co-infection among patients with pemphigus vulgaris. This retrospective study included 62 PV patients and 59 healthy individuals who attended the Second Affiliated Hospital of Kunming Medical University from November 2014 to November 2022. The serum concentrations of cytokines and chemokines were assessed using the Luminex 200 System (a high-throughput cytokine detection method). Additionally, anti-Dsg1 and anti-Dsg3 antibodies were determined through enzyme-linked immunosorbent assay, while disease severity was evaluated using the PDAI scoring system. The PV group exhibited elevated levels of Th1 cytokines (such as interleukin (IL)-1RA, IL-1ß, IL-2, IL-12p70, GM-CSF, TNF-α, IL-18, IFN-γ), Th2 cytokines (IL-5, IL-10, IL-13) and Th17/Th22-related cytokines (IL-17A, IL-22) compared to the healthy control group (p < 0.05). Conversely, the levels of chemokines (macrophage inflammatory protein-1 alpha (MIP-1α), stromal cell-derived factor-1 alpha (SDF-1α), interferon-inducible protein-10 (IP-10), Regulated on Activation in Normal T-Cell Expressed And Secreted (RANTES), growth-regulated on-gene-alpha (GRO-α), MIP-1ß) and Th2 (IL-31) were lower in the PV group compared to the healthy control group (p < 0.05). No significant differences were observed in other cytokines and chemokines (p > 0.05). Additionally, IL-7, IFN-γ, IL-18 and GRO-α showed positive correlations with PDAI, IL-6 correlated positively with anti-Dsg3 antibody levels, and IL-12p70, IL-18, and IFN-γ correlated positively with anti-Dsg1 antibody levels. Furthermore, IL-15 exhibited a positive association with skin infections. PV patients have elevated levels of various cytokines and chemokines, and there are different degrees of elevation in cytokines and chemokines associated with the activation of various T cell subsets. PDAI and the Dsg1 antibody levels are mainly related to the Th1-related cytokines.


Subject(s)
Chemokines , Cytokines , Desmoglein 1 , Pemphigus , Humans , Pemphigus/blood , Pemphigus/immunology , Retrospective Studies , Male , Female , Cytokines/blood , Middle Aged , Adult , Desmoglein 1/immunology , Chemokines/blood , Desmoglein 3/immunology , Severity of Illness Index , Aged , Autoantibodies/blood , Case-Control Studies , Clinical Relevance
11.
Cells ; 13(17)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39272976

ABSTRACT

3D bioprinting has become a valuable tool for studying the biology of solid tumors, including glioblastoma multiforme (GBM). Our analysis of publicly available bulk RNA and single-cell sequencing data has allowed us to define the chemotactic profile of GBM tumors and identify the cell types that secrete particular chemokines in the GBM tumor microenvironment (TME). Our findings indicate that primary GBM tissues express multiple chemokines, whereas spherical monocultures of GBM cells significantly lose this diversity. Subsequently, the comparative analysis of GBM spherical monocultures vs. 3D-bioprinted multicultures of cells showed a restoration of chemokine profile diversity in 3D-bioprinted cultures. Furthermore, single-cell RNA-Seq analysis showed that cells of the perivascular niche (pericytes and endocytes) express multiple chemokines in the GBM TME. Next, we 3D-bioprinted cells from two glioblastoma cell lines, U-251 and DK-MG, alone and as co-cultures with mesenchymal stromal cells (representing cells of the perivascular niche) and assessed the chemokine secretome. The results clearly demonstrated that the interaction of tumors and mesenchymal cells leads to in a significant increase in the repertoire and levels of secreted chemokines under culture in 21% O2 and 1% O2. Our study indicates that cells of the perivascular niche may perform a substantial role in shaping the chemokine microenvironment in GBM tumors.


Subject(s)
Chemokines , Coculture Techniques , Glioblastoma , Mesenchymal Stem Cells , Tumor Microenvironment , Humans , Glioblastoma/pathology , Glioblastoma/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Chemokines/metabolism , Cell Line, Tumor , Printing, Three-Dimensional , Bioprinting , Brain Neoplasms/pathology , Brain Neoplasms/metabolism
12.
Cancer Immunol Immunother ; 73(11): 229, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39249543

ABSTRACT

Immune checkpoint inhibitors are approved for recurrent/metastatic head and neck squamous cell carcinoma (HNSCC) but the response rate is only 13-18%. For an effective antitumor immune response, trafficking of immune cells to the tumor microenvironment (TME) is essential. We aimed to better understand immune cell migration as well as the involved chemokines in HNSCC. A transwell assay was used to study immune cell migration toward TME-conditioned medium. While T cell migration was not observed, conventional dendritic cell (cDC) migration was induced by TME-conditioned media. cDC migration correlated with various proteins in the TME secretome. CCL8, CXCL5, CCL13 and CCL7 were tested in validation experiments and addition of these chemokines induced cDC migration. Using single cell RNA-sequencing, we observed expression of CCL8, CXCL5, CCL13 and CCL7 in cancer-associated fibroblasts (CAFs). Depleting fibroblasts led to reduced cDC migration. Thus CAFs, while often seen as suppressors of antitumor immunity, play a role in attracting cDCs toward the head and neck cancer TME, which might be crucial for effective antitumor immunity and response to therapies. Indeed, we found RNA expression signatures of the indicated chemokines, cDC and CAF subpopulations, to be significantly higher in baseline tumor specimen of patients with a major pathological response to pre-surgical anti-PD-1 treatment compared to non-responding patients.


Subject(s)
Cell Movement , Dendritic Cells , Head and Neck Neoplasms , Tumor Microenvironment , Humans , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Tumor Microenvironment/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Secretome/metabolism , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/immunology , Chemokines/metabolism
13.
Int J Mol Sci ; 25(17)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39273432

ABSTRACT

Dimethyl fumarate (DMF), originally proposed to treat multiple sclerosis, is considered to have a spectrum of anti-inflammatory effects that effectively control periodontitis, mainly when applied with a hydrogel delivery system. Chemokine expression by gingival fibroblasts is a significant driver of periodontitis; thus, hydrogel-based strategies to deliver DMF, which in turn dampen chemokine expression, are of potential clinical relevance. To test this approach, we have established a bioassay where chemokine expression is induced by exposing gingival fibroblast to IL1ß and TNFα, or with saliva. We show herein that DMF effectively reduced the expression of CXCL8, CXCL1, CXCL2, and CCL2-and lowered the phosphorylation of ERK and JNK-without affecting cell viability. This observation was confirmed by immunoassays with CXCL8. Consistently, the forced chemokine expression in HSC2 oral squamous epithelial cells was greatly diminished by DMF. To implement our hydrogel-based delivery system, gingival fibroblasts were cocultured with gellan gum hydrogels enriched for DMF. In support of our strategy, DMF-enriched gellan gum hydrogels significantly reduced the forced chemokine expression in gingival fibroblasts. Our data suggest that DMF exerts its anti-inflammatory activity in periodontal cells when released from gellan gum hydrogels, suggesting a potential clinical relevance to control overshooting chemokine expression under chronic inflammatory conditions.


Subject(s)
Chemokines , Dimethyl Fumarate , Fibroblasts , Gingiva , Hydrogels , Polysaccharides, Bacterial , Humans , Hydrogels/chemistry , Dimethyl Fumarate/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Chemokines/metabolism , Gingiva/cytology , Gingiva/metabolism , Polysaccharides, Bacterial/pharmacology , Polysaccharides, Bacterial/chemistry , Cell Survival/drug effects , Cell Line
14.
Front Immunol ; 15: 1447161, 2024.
Article in English | MEDLINE | ID: mdl-39267759

ABSTRACT

Introduction: Tuberculosis (TB) remains a significant health concern in India, and its complexity is exacerbated by the rising occurrence of non-communicable diseases such as diabetes mellitus (DM). Recognizing that DM is a risk factor for active TB, the emerging comorbidity of TB and PDM (TB-PDM) presents a particular challenge. Our study focused on the impact of PDM on cytokine and chemokine profiles in patients with pulmonary tuberculosis TB) who also have PDM. Materials and methods: We measured and compared the cytokine (GM-CSF, IFN-γ, IL-1α/IL-1F1, IL-1ß/IL-1F2, IL-2, IL-4, IL-5, IL-6, IL-10, IL-12p70, IL-13, IL-17/IL-17A, IL-18/IL-1F4, TNF-α) and chemokine (CCL1, CCL2, CCL3, CCL4, CCL11, CXCL1, CXCL2, CXCL9, CXCL10, and CXCL11) levels in plasma samples of TB-PDM, only TB or only PDM using multiplex assay. Results: We observed that PDM was linked to higher mycobacterial loads in TB. Patients with coexisting TB and PDM showed elevated levels of various cytokines (including IFNγ, TNFα, IL-2, IL-17, IL-1α, IL-1ß, IL-6, IL-12, IL-18, and GM-CSF) and chemokines (such as CCL1, CCL2, CCL3, CCL4, CCL11, CXCL1, CXCL9, CXCL10, and CXCL11). Additionally, cytokines such as IL-18 and GM-CSF, along with the chemokine CCL11, were closely linked to levels of glycated hemoglobin (HbA1c), hinting at an interaction between glycemic control and immune response in TB patients with PDM. Conclusion: Our results highlight the complex interplay between metabolic disturbances, immune responses, and TB pathology in the context of PDM, particularly highlighting the impact of changes in HbA1c levels. This emphasizes the need for specialized approaches to manage and treat TB-PDM comorbidity.


Subject(s)
Cytokines , Prediabetic State , Tuberculosis, Pulmonary , Humans , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/blood , Cytokines/blood , Male , Female , Adult , Middle Aged , Prediabetic State/immunology , Prediabetic State/blood , Chemokines/blood , Biomarkers/blood , Mycobacterium tuberculosis/immunology , India/epidemiology
16.
Clin Cancer Res ; 30(19): 4434-4449, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39101835

ABSTRACT

PURPOSE: Clinical efficacy of chimeric antigen receptor (CAR) T cells against pediatric osteosarcoma (OS) has been limited. One strategy to improve efficacy may be to drive chemokine-mediated homing of CAR T cells to tumors. We sought to determine the primary chemokines secreted by OS and evaluate the efficacy of B7-H3.CAR T cells expressing the cognate receptors. EXPERIMENTAL DESIGN: We developed a pipeline to identify chemokines secreted by OS by correlating RNA-seq data with chemokine protein detected in media from fresh surgical specimens. We identified CXCR2 and CXCR6 as promising receptors for enhancing CAR T-cell homing against OS. We evaluated the homing kinetics and efficiency of CXCR2- and CXCR6.T cells and homing, cytokine production, and antitumor activity of CXCR2- and CXCR6.B7-H3.CAR T cells in vitro and in vivo. RESULTS: T cells transgenically expressing CXCR2 or CXCR6 exhibited ligand-specific enhanced migration over T cells modified with nonfunctional control receptors. Differential homing kinetics were observed, with CXCR2.T-cell homing quickly and plateauing early, whereas CXCR6.T cells took longer to home but achieved a similar plateau. When expressed in B7-H3.CAR T cells, CXCR2- and CXCR6 modification conferred enhanced homing toward OS in vitro and in vivo. CXCR2- and CXCR6-B7-H3.CAR-treated mice experienced prolonged survival in a metastatic model compared with B7-H3.CAR T-cell-treated mice. CONCLUSIONS: Our patient-based pipeline identified targets for chemokine receptor modification of CAR T cells targeting OS. CXCR2 and CXCR6 expression enhanced the homing and anti-OS activity of B7-H3.CAR T cells. These findings support clinical evaluation of CXCR-modified CAR T cells to improve adoptive cell therapy for patients with OS.


Subject(s)
B7 Antigens , Chemokines , Immunotherapy, Adoptive , Osteosarcoma , Receptors, CXCR6 , Receptors, Chimeric Antigen , Xenograft Model Antitumor Assays , Osteosarcoma/immunology , Osteosarcoma/therapy , Osteosarcoma/pathology , Osteosarcoma/genetics , Animals , Humans , Mice , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Receptors, CXCR6/genetics , Receptors, CXCR6/metabolism , Receptors, CXCR6/immunology , B7 Antigens/genetics , B7 Antigens/metabolism , Chemokines/metabolism , Cell Line, Tumor , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Cell Movement
17.
Front Immunol ; 15: 1404373, 2024.
Article in English | MEDLINE | ID: mdl-39114667

ABSTRACT

Background: Many cancers metastasize to the pleura, resulting in effusions that cause dyspnea and discomfort. Regardless of the tissue of origin, pleural malignancies are aggressive and uniformly fatal, with no treatment shown to prolong life. The pleural mesothelial monolayer is joined by tight junctions forming a contained bioreactor-like space, concentrating cytokines and chemokines secreted by the mesothelium, tumor, and infiltrating immune cells. This space represents a unique environment that profoundly influences tumor and immune cell behavior. Defining the pleural secretome is an important step in the rational development localized intrapleural immunotherapy. Method: We measured cytokine/chemokine content of 252 malignant pleural effusion (MPE) samples across multiple cancers using a 40-analyte panel and Luminex multiplexing technology. Results: Eleven analytes were consistently present in concentrations ≥ 10.0 pM: CXCL10/IP10 (geometric mean = 672.3 pM), CCL2/MCP1 (562.9 pM), sIL-6Rα (403.1 pM), IL-6 (137.6 pM), CXCL1/GRO (80.3 pM), TGFß1 (76.8 pM), CCL22/MDC (54.8 pM), CXCL8/IL-8 (29.2 pM), CCL11/Eotaxin (12.6 pM), IL-10 (11.3 pM), and G-CSF (11.0 pM). All are capable of mediating chemotaxis, promotion of epithelial to mesenchymal transition, or immunosuppression, and many of are reportedly downstream of a pro-inflammatory cytokine cascade mediated by cytokine IL-6 and its soluble receptor. Conclusion: The data indicate high concentrations of several cytokines and chemokines across epithelial cancers metastatic to the pleura and support the contention that the pleural environment is the major factor responsible for the clinical course of MPE across cancer types. A sIL-6Rα to IL-6 molar ratio of 2.7 ensures that virtually all epithelial, immune and vascular endothelial cells in the pleural environment are affected by IL-6 signaling. The central role likely played by IL-6 in the pathogenesis of MPE argues in favor of a therapeutic approach targeting the IL-6/IL-6R axis.


Subject(s)
Interleukin-6 , Pleural Effusion, Malignant , Humans , Pleural Effusion, Malignant/metabolism , Pleural Effusion, Malignant/pathology , Pleural Effusion, Malignant/immunology , Interleukin-6/metabolism , Pleural Neoplasms/metabolism , Pleural Neoplasms/pathology , Pleural Neoplasms/immunology , Pleural Neoplasms/secondary , Cytokines/metabolism , Neoplasms, Glandular and Epithelial/pathology , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/immunology , Female , Male , Aged , Biomarkers, Tumor/metabolism , Chemokines/metabolism , Signal Transduction , Tumor Microenvironment/immunology , Middle Aged
18.
Front Immunol ; 15: 1443366, 2024.
Article in English | MEDLINE | ID: mdl-39114657

ABSTRACT

The tumor microenvironment (TME) is a complex interconnected network of immune cells, fibroblasts, blood vessels, and extracellular matrix surrounding the tumor. Because of its immunosuppressive nature, the TME can pose a challenge for cancer immunotherapies targeting solid tumors. Chemokines have emerged as a crucial element in enhancing the efficacy of cancer immunotherapy, playing a direct role in immune cell signaling within the TME and facilitating immune cell migration towards cancer cells. However, chemokine ligands and their receptors exhibit context-dependent diversity, necessitating evaluation of their tumor-promoting or inhibitory effects based on tumor type and immune cell characteristics. This review explores the role of chemokines in tumor immunity and metastasis in the context of the TME. We also discuss current chemokine-related advances in cancer immunotherapy research, with a particular focus on lung cancer, a common cancer with a low survival rate and limited immunotherapy options.


Subject(s)
Chemokines , Immunotherapy , Lung Neoplasms , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Lung Neoplasms/pathology , Immunotherapy/methods , Chemokines/metabolism , Chemokines/immunology , Animals , Signal Transduction
19.
Pharmacol Rep ; 76(5): 1174-1183, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39088104

ABSTRACT

BACKGROUND: - Alcohol-induced pro-inflammatory activation might influence cellular and synaptic pathology, thus contributing to the behavioral phenotypes associated with alcohol use disorders. In the present study, the possible anti-inflammatory properties of N-[(4-trifluoromethyl)-benzyl]4-methoxybutyramide (GET73), a promising therapeutic agent for alcohol use disorder treatment, were evaluated in primary cultures of rat cortical microglia. METHODS: - Primary cultures of cerebral cortex microglial cells were treated with 100 ng/ml lipopolysaccharide (LPS; 8 h, 37 °C) or 75 mM ethanol (EtOH; 4 days, 37 °C) alone or in the presence of GET73 (1-30 µM). At the end of the incubation period, multiparametric quantification of cytokines/chemokines was performed by using the xMAP technology and Luminex platform. Furthermore, cultured microglial cell viability following the treatment with EtOH and GET73, alone or in combination, has been measured by a colorimetric assay (i.e. MTT assay). RESULTS: - GET73 (10 and 30 µM) partially or fully prevented the LPS-induced increase of IL-6, IL-1ß, RANTES/CCL5 protein and MCP-1/CCL2 levels. On the contrary, GET73 failed to attenuate the TNF-α level increase induced by LPS. Furthermore, GET73 treatment (10-30 µM) significantly attenuated or prevented the EtOH-induced increase of TNF-α, IL-6, IL-1ß and MCP-1/CCL2 levels. Finally, at all the concentrations tested (1-30 µM), the GET73 treatment did not alter the EtOH-induced reduction of microglial cell viability. CONCLUSIONS: - The current results provide the first in vitro evidence of GET73 protective properties against EtOH-induced neuroinflammation. These data add more information on the complex and multifactorial profile of action of the compound, further supporting the significance of developing GET73 as a therapeutic tool for the treatment of individuals with alcohol use disorders.


Subject(s)
Cell Survival , Cerebral Cortex , Cytokines , Ethanol , Lipopolysaccharides , Microglia , Animals , Rats , Anti-Inflammatory Agents/pharmacology , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Chemokines/metabolism , Cytokines/metabolism , Ethanol/pharmacology , Lipopolysaccharides/pharmacology , Microglia/drug effects , Microglia/metabolism , Rats, Wistar
20.
Front Immunol ; 15: 1456405, 2024.
Article in English | MEDLINE | ID: mdl-39206194

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common primary neoplasms of the liver and one of the most common solid tumors in the world. Its global incidence is increasing and it has become the third leading cause of cancer-related deaths. There is growing evidence that chemokines play an important role in the tumor microenvironment, regulating the migration and localization of immune cells in tissues and are critical for the function of the immune system. This review comprehensively analyses the expression and activity of chemokines in the TME of HCC and describes their interrelationship with hepatocarcinogenesis and progression. Special attention is given to the role of chemokine-chemokine receptors in the regulation of immune cell accumulation in the TME. Therapeutic strategies targeting tumor-promoting chemokines or the induction/release of beneficial chemokines are reviewed, highlighting the potential value of natural products in modulating chemokines and their receptors in the treatment of HCC. The in-depth discussion in this paper provides a theoretical basis for the treatment of HCC. It is an important reference for new drug development and clinical research.


Subject(s)
Biological Products , Carcinoma, Hepatocellular , Chemokines , Liver Neoplasms , Tumor Microenvironment , Humans , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/immunology , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Tumor Microenvironment/immunology , Chemokines/metabolism , Biological Products/therapeutic use , Biological Products/pharmacology , Animals , Receptors, Chemokine/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL