Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.325
Filter
1.
Med Sci Monit ; 30: e943170, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39018268

ABSTRACT

BACKGROUND Tumor-infiltrating immune cells (TIICs) are implicated in the survival of ovarian cancer (OVCA) patients, but their prognostic significance in advanced or metastatic OVCA patients treated with neoadjuvant chemotherapy (NCAT) has not been well documented, particularly in the Chinese population. MATERIAL AND METHODS A total of 31 advanced or metastatic OVCA patients who underwent NACT were included. The density and positive rate of tumor-infiltrating immune cells (TIICs) within cancer cell nests and in cancer stroma were explored. The correlations of pre- or post-NACT TIICs with the efficacy of NACT and the changes in TIIC subpopulation with NACT were examined. RESULTS Compared with patients with partial benefit from NACT, significantly decreased pre-NACT intratumoral CD68⁺CD163⁺ cells (P=0.0043) and increased pre-NACT intratumoral CD56⁺ cells (P=0.038) were observed in patients with benefit. The high level of pre-NACT intratumoral CD68⁺CD163⁻ M1 macrophage (P=0.075) and stromal CD3⁺PD-1⁺ cells (P=0.085) predicated improved progression-free survival, respectively. Increased post-NACT stromal CD68⁺CD163⁻ M1 macrophage (P=0.01), stromal CD8⁺ T cells (P=0.073), and stromal CD8⁺PD-1⁺ cells (P=0.072) were associated with benefit from NACT. Moreover, NACT increased intratumoral CD3⁺ (P=0.031), CD8+ (P=0.031), and CD3⁺CD8⁺ cells (P=0.031). CONCLUSIONS High intratumoral CD68⁺CD163⁻, intratumoral CD56⁺ cells, and stromal CD3⁺PD-1⁺ cells pre-NACT predicted good prognosis. Intratumoral CD3⁺, CD8⁺, and CD3⁺CD8⁺ cells were increased after NACT. Evaluation of immune profiles may help to identify patients who might benefit from NACT and allow us to further stratify advanced or metastatic OVCA patients treated with NACT for disease management.


Subject(s)
Lymphocytes, Tumor-Infiltrating , Neoadjuvant Therapy , Ovarian Neoplasms , Humans , Female , Neoadjuvant Therapy/methods , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Middle Aged , Prognosis , Retrospective Studies , Lymphocytes, Tumor-Infiltrating/immunology , Adult , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Neoplasm Metastasis , Treatment Outcome , Tumor Microenvironment/immunology , China , Receptors, Cell Surface
2.
J Reprod Immunol ; 164: 104296, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38972269

ABSTRACT

Preeclampsia is a disorder of pregnancy characterized by endothelial dysfunction, abnormal placentation, systemic inflammation, and altered immune reaction. The aim of this study was to investigate the immune checkpoint molecules TIM-3 and Gal-9 on macrophages and Hofbauer cells (HBC) in the placenta of preeclampsia patients. Immunohistochemistry and Immunofluorescence was used to characterize the expression of the macrophage markers CD68 and CD163, CK7 and the proteins TIM-3 and Gal-9 in the placentas of preeclampsia patients comparing it to the placentas of healthy pregnancies. Double immunofluorescence staining (TIM-3 with CD3/CD19/CD56) was used to analyze the TIM-3 expression on other immune cells (T cells, B cells, NK cells) within the chorionic villi. The expression of TIM-3 on decidual macrophages did not significantly differ between the preeclamptic and the control group (p = 0.487). When looking at the different offspring we saw an upregulation of TIM-3 expression on decidual macrophages in preeclamptic placentas with female offspring (p = 0.049). On Hofbauer cells within the chorionic villi, the TIM-3 expression was significantly downregulated in preeclamptic cases without a sex-specific difference (p < 0.001). Looking at the protein Gal-9 the expression was proven to be downregulated both, on decidual macrophages (p = 0.003) and on Hofbauer cells (p = 0.002) within preeclamptic placentas compared to healthy controls. This was only significant in male offspring (p < 0.001 and p = 0.013) but not in female offspring (p = 0.360 and p = 0.068). While TIM-3 expression within the extravillious trophoblast and the syncytiotrophoblast was significantly downregulated (p < 0.001 and p = 0.012) in preeclampsia, the expression of Gal-9 was upregulated in (p < 0.001 and p < 0.001) compared to healthy controls. The local variations of the immune checkpoint molecules TIM-3 and Gal-9 in the placenta may contribute to the inflammation observed in preeclamptic patients. It could therefore contribute to the pathogenesis and be an important target in the treatment of preeclampsia.


Subject(s)
Galectins , Hepatitis A Virus Cellular Receptor 2 , Macrophages , Placenta , Pre-Eclampsia , Humans , Pregnancy , Hepatitis A Virus Cellular Receptor 2/metabolism , Female , Pre-Eclampsia/immunology , Pre-Eclampsia/metabolism , Pre-Eclampsia/pathology , Macrophages/immunology , Macrophages/metabolism , Galectins/metabolism , Adult , Placenta/immunology , Placenta/metabolism , Placenta/pathology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Receptors, Cell Surface/metabolism , Chorionic Villi/immunology , Chorionic Villi/metabolism , Chorionic Villi/pathology , Male
3.
J Transl Med ; 22(1): 674, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39039496

ABSTRACT

BACKGROUND: Preeclampsia, especially early-onset preeclampsia (EO-PE), is a pregnancy complication that has serious consequences for the health of both the mother and the fetus. Although abnormal placentation due to mitochondrial dysfunction is speculated to contribute to the development of EO-PE, the underlying mechanisms have yet to be fully elucidated. METHODS: The expression and localization of Siglec-6 in the placenta from normal pregnancies, preterm birth and EO-PE patients were examined by RT-qPCR, Western blot and IHC. Transwell assays were performed to evaluate the effect of Siglec-6 on trophoblast cell migration and invasion. Seahorse experiments were conducted to assess the impact of disrupting Siglec-6 expression on mitochondrial function. Co-IP assay was used to examine the interaction of Siglec-6 with SHP1/SHP2. RNA-seq was employed to investigate the mechanism by which Siglec-6 inhibits mitochondrial function in trophoblast cells. RESULTS: The expression of Siglec-6 in extravillous trophoblasts is increased in placental tissues from EO-PE patients. Siglec-6 inhibits trophoblast cell migration and invasion and impairs mitochondrial function. Mechanismly, Siglec-6 inhibits the activation of NF-κB by recruiting SHP1/SHP2, leading to increased expression of GPR20. Notably, the importance of GPR20 function downstream of Siglec-6 in trophoblasts is supported by the observation that GPR20 downregulation rescues defects caused by Siglec-6 overexpression. Finally, overexpression of Siglec-6 in the placenta induces a preeclampsia-like phenotype in a pregnant mouse model. CONCLUSIONS: This study indicates that the regulatory pathway Siglec-6/GPR20 has a crucial role in regulating trophoblast mitochondrial function, and we suggest that Siglec-6 and GPR20 could serve as potential markers and targets for the clinical diagnosis and therapy of EO-PE.


Subject(s)
Cell Movement , Mitochondria , Pre-Eclampsia , Receptors, G-Protein-Coupled , Trophoblasts , Up-Regulation , Pre-Eclampsia/metabolism , Pre-Eclampsia/genetics , Pre-Eclampsia/pathology , Humans , Pregnancy , Female , Mitochondria/metabolism , Up-Regulation/genetics , Trophoblasts/metabolism , Animals , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Cell Movement/genetics , Lectins/metabolism , Placenta/metabolism , Mice , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Adult
4.
Sci Rep ; 14(1): 14974, 2024 06 28.
Article in English | MEDLINE | ID: mdl-38951667

ABSTRACT

Bovine alveolar macrophages (AMs) defend the lungs against pathogens such as Mycobacterium bovis (M. bovis), the causative agent of bovine tuberculosis. However, little is known about the surface molecules expressed by bovine AMs and whether there is heterogeneity within the population. The purpose of this study was to characterise the bovine AM cell surface phenotype using flow cytometry. Bronchoalveolar lavage samples from four different calves were stained with a combination of antibodies against immune cell molecules prior to flow cytometric analysis. To assess the degree of expression, we considered the distribution and relative intensities of stained and unstained cells. We demonstrated that bovine AMs have high expression of CD172a, ADGRE1, CD206, and CD14, moderate expression of CD80, MHC II, CD1b, and CD40, low expression of CX3CR1 and CD86, and little or no expression of CD16 and CD26. Two distinct subsets of bovine AMs were identified based on CD163 expression. Subsequent analysis showed that the CD163+ subset had greater expression of other typical macrophage molecules compared to the CD163- subset, suggesting that these cells may perform different roles during infection. The characterisation of the uninfected bovine AM phenotype will provide a foundation for the examination of M. bovis-infected AMs.


Subject(s)
Antigens, CD , Antigens, Differentiation, Myelomonocytic , Macrophages, Alveolar , Receptors, Cell Surface , Animals , Cattle , Macrophages, Alveolar/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, CD/metabolism , Receptors, Cell Surface/metabolism , Phenotype , Mycobacterium bovis/immunology , Flow Cytometry , Tuberculosis, Bovine/metabolism , Tuberculosis, Bovine/immunology , Tuberculosis, Bovine/microbiology , Immunophenotyping , Bronchoalveolar Lavage Fluid
5.
J Nanobiotechnology ; 22(1): 388, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956618

ABSTRACT

BACKGROUND: Porcine reproductive and respiratory syndrome virus (PRRSV) is a prevalent swine pathogen, which has caused adverse impact on the global swine industry for almost 30 years. However, due to the immune suppression caused by the virus and the genetic diversity in PRRSV, no virus-targeting broad neutralizing strategy has been successfully developed yet. Antiviral peptide and nanobody have attracted extensive attention with the ease in production and the efficacy in practice. In this study, four new fusion proteins named nanobody peptide conjugates (NPCs) were developed by combining PRRSV specific non-neutralizing nanobodies with CD163-derived peptides targeting the receptor binding domain (RBD) of PRRSV proteins. RESULTS: Four NPCs were successfully constructed using two nanobodies against PRRSV N and nsp9 individually, recombining with two antiviral peptides 4H7 or 8H2 from porcine CD163 respectively. All four NPCs demonstrated specific capability of binding to PRRSV and broad inhibitory effect against various lineages of PRRSV in a dose-dependent manner. NPCs interfere with the binding of the RBD of PRRSV proteins to CD163 in the PRRSV pre-attachment stage by CD163 epitope peptides in the assistance of Nb components. NPCs also suppress viral replication during the stage of post-attachment, and the inhibitory effects depend on the antiviral functions of Nb parts in NPCs, including the interference in long viral RNA synthesis, NF-κB and IFN-ß activation. Moreover, an interaction was predicted between aa K31 and T32 sites of neutralizing domain 4H7 of NPC-N/nsp9-4H7 and the motif 171NLRLTG176 of PRRSV GP2a. The motif 28SSS30 of neutralizing domain 8H2 of NPC-N/nsp9-8H2 could also form hydrogens to bind with the motif 152NAFLP156 of PRRSV GP3. The study provides valuable insights into the structural characteristics and potential functional implications of the RBD of PRRSV proteins. Finally, as indicated in a mouse model, NPC intranasally inoculated in vivo for 12-24 h sustains the significant neutralizing activity against PRRSV. These findings inspire the potential of NPC as a preventive measure to reduce the transmission risk in the host population against respiratory infectious agents like PRRSV. CONCLUSION: The aim of the current study was to develop a peptide based bioactive compound to neutralize various PRRSV strains. The new antiviral NPC (nanobody peptide conjugate) consists of a specific nanobody targeting the viral protein and a neutralizing CD163 epitope peptide for virus blocking and provides significant antiviral activity. The study will greatly promote the antiviral drug R&D against PRRSV and enlighten a new strategy against other viral diseases.


Subject(s)
Antibodies, Neutralizing , Antigens, CD , Antigens, Differentiation, Myelomonocytic , Peptides , Porcine respiratory and reproductive syndrome virus , Receptors, Cell Surface , Single-Domain Antibodies , Porcine respiratory and reproductive syndrome virus/immunology , Porcine respiratory and reproductive syndrome virus/drug effects , Animals , Single-Domain Antibodies/immunology , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/chemistry , Swine , Antigens, Differentiation, Myelomonocytic/immunology , Antigens, Differentiation, Myelomonocytic/metabolism , Receptors, Cell Surface/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Antibodies, Neutralizing/immunology , Peptides/chemistry , Peptides/pharmacology , Peptides/immunology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/prevention & control , Mice , Virus Replication/drug effects , Cell Line
6.
Eur J Med Res ; 29(1): 357, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970071

ABSTRACT

BACKGROUND AND PURPOSE: PD-1/PD-L1 inhibitors have become a promising therapy. However, the response rate is lower than 30% in patients with cervical cancer (CC), which is related to immunosuppressive components in tumor microenvironment (TME). Tumor-associated macrophages (TAMs), as one of the most important immune cells, are involved in the formation of tumor suppressive microenvironment. Therefore, it will provide a theoretical basis for curative effect improvement about the regulatory mechanism of TAMs on PD-L1 expression. METHODS: The clinical data and pathological tissues of CC patients were collected, and the expressions of PD-L1, CD68 and CD163 were detected by immunohistochemistry. Bioinformatics was used to analyze the macrophage subtypes involved in PD-L1 regulation. A co-culture model was established to observe the effects of TAMs on the morphology, migration and invasion function of CC cells, and the regulatory mechanism of TAMs on PD-L1. RESULTS: PD-L1 expression on tumor cells could predict the poor prognosis of patients. And there was a strong correlation between PD-L1 expression with CD163+TAMs infiltration. Similarly, PD-L1 expression was associated with M1/M2-type TAMs infiltration in bioinformatics analysis. The results of cell co-culture showed that M1/M2-type TAMs could upregulate PD-L1 expression, especially M2-type TAMs may elevate the PD-L1 expression via PI3K/AKT pathway. Meanwhile, M1/M2-type TAMs can affect the morphological changes, and enhance migration and invasion abilities of CC cells. CONCLUSIONS: PD-L1 expression in tumor cells can be used as a prognostic factor and is closely related to CD163+TAMs infiltration. In addition, M2-type TAMs can upregulate PD-L1 expression in CC cells through PI3K/AKT pathway, enhance the migration and invasion capabilities, and affect the tumor progression.


Subject(s)
B7-H1 Antigen , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Tumor-Associated Macrophages , Uterine Cervical Neoplasms , Humans , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/immunology , Female , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Tumor Microenvironment/immunology , Up-Regulation , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Middle Aged , Antigens, CD/metabolism , Antigens, CD/genetics , Prognosis , Gene Expression Regulation, Neoplastic , Cell Movement , Receptors, Cell Surface
7.
Zool Res ; 45(4): 833-844, 2024 07 18.
Article in English | MEDLINE | ID: mdl-39004861

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) is a globally prevalent contagious disease caused by the positive-strand RNA PRRS virus (PRRSV), resulting in substantial economic losses in the swine industry. Modifying the CD163 SRCR5 domain, either through deletion or substitution, can eff1ectively confer resistance to PRRSV infection in pigs. However, large fragment modifications in pigs inevitably raise concerns about potential adverse effects on growth performance. Reducing the impact of genetic modifications on normal physiological functions is a promising direction for developing PRRSV-resistant pigs. In the current study, we identified a specific functional amino acid in CD163 that influences PRRSV proliferation. Viral infection experiments conducted on Marc145 and PK-15 CD163 cells illustrated that the mE535G or corresponding pE529G mutations markedly inhibited highly pathogenic PRRSV (HP-PRRSV) proliferation by preventing viral binding and entry. Furthermore, individual viral challenge tests revealed that pigs with the E529G mutation had viral loads two orders of magnitude lower than wild-type (WT) pigs, confirming effective resistance to HP-PRRSV. Examination of the physiological indicators and scavenger function of CD163 verified no significant differences between the WT and E529G pigs. These findings suggest that E529G pigs can be used for breeding PRRSV-resistant pigs, providing novel insights into controlling future PRRSV outbreaks.


Subject(s)
Antigens, CD , Antigens, Differentiation, Myelomonocytic , Point Mutation , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Receptors, Cell Surface , Animals , Swine , Porcine Reproductive and Respiratory Syndrome/genetics , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/physiology , Porcine respiratory and reproductive syndrome virus/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Animals, Genetically Modified/genetics , Cell Line
8.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1126-1134, 2024 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-38977342

ABSTRACT

OBJECTIVE: To investigate the protective effect of recombinant Schistosoma japonicum cystatin (rSj-Cys) against acute liver injury induced by lipopolysaccharide (LPS) and D-GalN in mice. METHODS: Adult male C57BL/6J mice with or without LPS/D-GaIN-induced acute liver injury were given intraperitoneal injections of rSj-Cys or PBS 30 min after modeling (n=18), and serum and liver tissues samples were collected from 8 mice in each group 6 h after modeling. The survival of the remaining 10 mice in each group within 24 h was observed. Serum levels of ALT, AST, TNF-α and IL-6 of the mice were measured, and liver pathologies was observed with HE staining. The hepatic expressions of macrophage marker CD68, Bax, Bcl-2 and endoplasmic reticulum stress (ERS)-related proteins were detected using immunohistochemistry or immunoblotting, and TUNEL staining was used to detect hepatocyte apoptosis. RESULTS: The survival rates of PBS- and rSj-Cys-treated mouse models of acute liver injury were 30% and 80% at 12 h and were 10% and 60% at 24 h after modeling, respectively; no death occurred in the two control groups within 24 h. The mouse models showed significantly increased serum levels of AST, ALT, IL-6 and TNF-α and serious liver pathologies with increased hepatic expressions of CD68 and Bax, lowered expression of Bcl-2, increased hepatocyte apoptosis, and up-regulated expressions of ERS-related signaling pathway proteins GRP78, CHOP and NF-κB p-p65. Treatment of the mouse models significantly lowered the levels of AST, ALT, IL-6 and TNF-α, alleviated liver pathologies, reduced hepatic expressions of CD68, Bax, GRP78, CHOP and NF-κB p-p65, and enhanced the expression of Bcl-2. In the normal control mice, rSj-Cys injection did not produce any significant changes in these parameters compared with PBS. CONCLUSION: rSj-Cys alleviates LPS/D-GalN-induced acute liver injury in mice by suppressing ERS, attenuating inflammation and inhibiting hepatocyte apoptosis.


Subject(s)
Apoptosis , Cystatins , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Hepatocytes , Inflammation , Mice, Inbred C57BL , Schistosoma japonicum , Animals , Mice , Endoplasmic Reticulum Stress/drug effects , Apoptosis/drug effects , Male , Hepatocytes/metabolism , Hepatocytes/drug effects , Cystatins/pharmacology , Liver/pathology , Liver/metabolism , Lipopolysaccharides , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism , Recombinant Proteins/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Galactosamine , Antigens, CD/metabolism , Chemical and Drug Induced Liver Injury/drug therapy , CD68 Molecule
9.
J Med Virol ; 96(7): e29819, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39030992

ABSTRACT

Pregnant women represent a high-risk population for Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection. The presence of SARS-CoV-2 has been reported in placenta from infected pregnant women, but whether the virus influences placenta immune response remains unclear. We investigated the properties of maternal-fetal interface macrophages (MFMs) in a cohort of unvaccinated women who contracted coronavirus disease 2019 (COVID-19) during their pregnancy. We reported an infiltration of CD163+ macrophages in placenta from COVID-19 women 19 whereas lymphoid compartment was not affected. Isolated MFMs exhibited nonpolarized activated signature (NOS2, IDO1, IFNG, TNF, TGFB) mainly in women infected during the second trimester of pregnancy. COVID-19 during pregnancy primed MFM to produce type I and III interferon response to SARS-CoV-2 (Wuhan and δ strains), that were unable to elicit this in MFMs from healthy pregnant women. COVID-19 also primed SARS-CoV-2 internalization by MFM in an angiotensin-converting enzyme 2-dependent manner. Activation and recall responses of MFMs were influenced by fetal sex. Collectively, these findings support a role for MFMs in the local immune response to SARS-CoV-2 infection, provide a basis for protective placental immunity in COVID-19, and highlight the interest of vaccination in pregnant women.


Subject(s)
COVID-19 , Macrophages , Placenta , Pregnancy Complications, Infectious , SARS-CoV-2 , Humans , Female , Pregnancy , COVID-19/immunology , COVID-19/virology , Placenta/immunology , Placenta/virology , Macrophages/immunology , Macrophages/virology , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/immunology , SARS-CoV-2/immunology , Adult , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Virus Internalization
10.
Circ Res ; 135(2): e4-e23, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38860377

ABSTRACT

BACKGROUND: Cell phenotype switching is increasingly being recognized in atherosclerosis. However, our understanding of the exact stimuli for such cellular transformations and their significance for human atherosclerosis is still evolving. Intraplaque hemorrhage is thought to be a major contributor to plaque progression in part by stimulating the influx of CD163+ macrophages. Here, we explored the hypothesis that CD163+ macrophages cause plaque progression through the induction of proapoptotic endothelial-to-mesenchymal transition (EndMT) within the fibrous cap. METHODS: Human coronary artery sections from CVPath's autopsy registry were selected for pathological analysis. Athero-prone ApoE-/- and ApoE-/-/CD163-/- mice were used for in vivo studies. Human peripheral blood mononuclear cell-induced macrophages and human aortic endothelial cells were used for in vitro experiments. RESULTS: In 107 lesions with acute coronary plaque rupture, 55% had pathological evidence of intraplaque hemorrhage in nonculprit vessels/lesions. Thinner fibrous cap, greater CD163+ macrophage accumulation, and a larger number of CD31/FSP-1 (fibroblast specific protein-1) double-positive cells and TUNEL (terminal deoxynucleotidyl transferase-dUTP nick end labeling) positive cells in the fibrous cap were observed in nonculprit intraplaque hemorrhage lesions, as well as in culprit rupture sections versus nonculprit fibroatheroma sections. Human aortic endothelial cells cultured with supernatants from hemoglobin/haptoglobin-exposed macrophages showed that increased mesenchymal marker proteins (transgelin and FSP-1) while endothelial markers (VE-cadherin and CD31) were reduced, suggesting EndMT induction. Activation of NF-κB (nuclear factor kappa ß) signaling by proinflammatory cytokines released from CD163+ macrophages directly regulated the expression of Snail, a critical transcription factor during EndMT induction. Western blot analysis for cleaved caspase-3 and microarray analysis of human aortic endothelial cells indicated that apoptosis was stimulated during CD163+ macrophage-induced EndMT. Additionally, CD163 deletion in athero-prone mice suggested that CD163 is required for EndMT and plaque progression. Using single-cell RNA sequencing from human carotid endarterectomy lesions, a population of EndMT was detected, which demonstrated significant upregulation of apoptosis-related genes. CONCLUSIONS: CD163+ macrophages provoke EndMT, which may promote plaque progression through fibrous cap thinning.


Subject(s)
Antigens, CD , Antigens, Differentiation, Myelomonocytic , Macrophages , Plaque, Atherosclerotic , Receptors, Cell Surface , Humans , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Animals , Antigens, CD/metabolism , Antigens, CD/genetics , Macrophages/metabolism , Macrophages/pathology , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Mice , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Male , Mice, Knockout, ApoE , Mice, Inbred C57BL , Apoptosis , Female , Epithelial-Mesenchymal Transition , Coronary Vessels/pathology , Coronary Vessels/metabolism
11.
Front Immunol ; 15: 1412076, 2024.
Article in English | MEDLINE | ID: mdl-38903497

ABSTRACT

Macrophages play essential roles in maintaining tissue homeostasis and immune defence. However, their extensive infiltration into tumours has been linked to adverse outcomes in multiple human cancers. Within the tumour microenvironment (TME), tumour-associated macrophages (TAMs) promote tumour growth and metastasis, making them prime targets for cancer immunotherapy. Recent single-cell analysis suggest that proliferating TAMs accumulate in human cancers, yet their origins and differentiation pathways remain uncertain. Here, we show that a subpopulation of CD163+ TAMs proliferates in situ within the TME of melanoma, lung cancer, and breast cancer. Consistent with their potential role in suppressing anti-tumour activities of T cells, CD163+ TAMs express a range of potent immunosuppressive molecules, including PD-L1, PD-L2, IL-10, and TGF-ß. Other phenotypic markers strongly suggested that these cells originate from CD14+ CCR2+ monocytes, a cell population believed to have minimal capacity for proliferation. However, we demonstrate in vitro that certain myelopoietic cytokines commonly available within the TME induce robust proliferation of human monocytes, especially the combination of interleukin 3 (IL-3) and Macrophage Colony-Stimulating Factor 1 (M-CSF). Monocytic cells cultured with these cytokines efficiently modulate T cell proliferation, and their molecular phenotype recapitulates that of CD163+ TAMs. IL-3-driven proliferation of monocytic cells can be completely blocked by IL-4, associated with the induction of CDKN1A, alongside the upregulation of transcription factors linked to dendritic cell function, such as BATF3 and IRF4. Taken together, our work suggests several novel therapeutic routes to reducing immunosuppressive TAMs in human tumours, from blocking chemokine-mediated recruitment of monocytes to blocking their proliferation.


Subject(s)
Cell Proliferation , Monocytes , Tumor Microenvironment , Tumor-Associated Macrophages , Humans , Monocytes/immunology , Monocytes/metabolism , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Neoplasms/immunology , Neoplasms/pathology , Antigens, CD/metabolism , Female , Macrophages/immunology , Macrophages/metabolism , Receptors, Cell Surface/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cytokines/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology
12.
BMC Cancer ; 24(1): 768, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926671

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a 'difficult-to-treat' entity. To forecast its prognosis, we introduced a new biomarker, SARIFA (stroma areactive invasion front areas), which are areas at the tumour invasion front lacking desmoplastic stroma reaction upon malignant invasion in the surrounding tissue, leading to direct contact between tumour cells and adipocytes. SARIFA showed its significance in gastric and colorectal carcinoma, revealing lipid metabolism alternations that promote tumour progression. METHODS: We reviewed the SARIFA status of 166 PDAC cases on all available H&E-stained tumour slides from archival Whipple-resection specimens. SARIFA positivity was defined as SARIFA detection in at least 66% of the available slides. To investigate alterations in tumour metabolism and microenvironment, we performed immunohistochemical staining for FABP4, CD36 and CD68. To verify and quantify a supposed delipidation of adipocytes, adipose tissue was digitally morphometrised. RESULTS: In total, 53 cases (32%) were classified as SARIFA positive and 113 (68%) as SARIFA negative. Patients with SARIFA-positive PDAC showed a significantly worse overall survival compared with SARIFA-negative cases (median overall survival: 11.0 months vs. 22.0 months, HR: 1.570 (1.082-2.278), 95% CI, p = 0.018), which was independent from other prognostic markers (p = 0.014). At the invasion front of SARIFA-positive PDAC, we observed significantly higher expression of FABP4 (p < 0.0001) and higher concentrations of CD68+ macrophages (p = 0.031) related to a higher risk of tumour progression. CD36 staining showed no significant expression differences. The adipocyte areas at the invasion front were significantly smaller, with mean values of 4021 ± 1058 µm2 and 1812 ± 1008 µm2 for the SARIFA-negative and -positive cases, respectively (p < 0.001). CONCLUSIONS: SARIFA is a promising prognostic biomarker for PDAC. Its assessment is characterised by simplicity and low effort. The mechanisms behind SARIFA suggest a tumour-promoting increased lipid metabolism and altered immune background, both showing new therapeutic avenues.


Subject(s)
Biomarkers, Tumor , Carcinoma, Pancreatic Ductal , Fatty Acid-Binding Proteins , Pancreatic Neoplasms , Humans , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Female , Male , Biomarkers, Tumor/metabolism , Prognosis , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Aged , Middle Aged , Fatty Acid-Binding Proteins/metabolism , Neoplasm Invasiveness , Tumor Microenvironment , Lipid Metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, CD/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology , CD36 Antigens/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adult , Aged, 80 and over , CD68 Molecule
13.
Cytokine ; 181: 156677, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38896955

ABSTRACT

BACKGROUND: Sepsis-associated encephalopathy (SAE) is a diffuse brain dysfunction activated by microglia. The potential pathological changes of SAE are complex, and the cellular pathophysiological characteristics remains unclear. This study aims to explore the ROS/TXNIP/NLRP3 pathway mediated lipopolysaccharide (LPS)-induced inflammatory response in microglia. METHODS: BV-2 cells were pre-incubated with 10 µM N-acetyl-L-cysteine (NAC) for 2 h, which were then reacted with 1 µg/mL LPS for 24 h. Western blot assay examined the protein levels of IBA1, CD68, TXNIP, NLRP3, ASC, and Cleaved Caspase-1 in BV-2 cells. The contents of inflammatory factor were detected by ELISA assay. The co-immunoprecipitation assay examined the interaction between TXNIP and NLRP3. RESULTS: LPS was confirmed to promote the positive expressions of IBA1 and CD68 in BV-2 cells. The further experiments indicated that LPS enhanced ROS production and NLRP3 inflammasome activation in BV-2 cells. Moreover, we also found that NAC partially reversed the facilitation of LPS on the levels of ROS, IL-1ß, IL-18, TXNIP, NLRP3, ASC, and Cleaved Caspase-1 in BV-2 cells. NAC treatment also notably alleviated the interaction between TXNIP and NLRP3 in BV-2 cells. CONCLUSION: ROS inhibition mediated NLRP3 signaling inactivation by decreasing TXNIP expression.


Subject(s)
Carrier Proteins , Caspase 1 , Inflammasomes , Inflammation , Lipopolysaccharides , Microglia , NLR Family, Pyrin Domain-Containing 3 Protein , Reactive Oxygen Species , Signal Transduction , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Microglia/metabolism , Microglia/drug effects , Lipopolysaccharides/pharmacology , Carrier Proteins/metabolism , Animals , Mice , Reactive Oxygen Species/metabolism , Caspase 1/metabolism , Signal Transduction/drug effects , Inflammasomes/metabolism , Inflammation/metabolism , Inflammation/pathology , Cell Line , Acetylcysteine/pharmacology , Calcium-Binding Proteins/metabolism , Interleukin-1beta/metabolism , Interleukin-18/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Microfilament Proteins/metabolism , Thioredoxins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Sepsis-Associated Encephalopathy/metabolism , Sepsis-Associated Encephalopathy/pathology , CD68 Molecule
14.
Medicina (Kaunas) ; 60(6)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38929614

ABSTRACT

Background and Objectives: Inflammatory proteins and their prognostic value in patients with carotid artery stenosis (CAS) have not been adequately studied. Herein, we identified CAS-specific biomarkers from a large pool of inflammatory proteins and assessed the ability of these biomarkers to predict adverse events in individuals with CAS. Materials and Methods: Samples of blood were prospectively obtained from 336 individuals (290 with CAS and 46 without CAS). Plasma concentrations of 29 inflammatory proteins were determined at recruitment, and the patients were followed for 24 months. The outcome of interest was a major adverse cardiovascular event (MACE; composite of stroke, myocardial infarction, or death). The differences in plasma protein concentrations between patients with vs. without a 2-year MACE were determined using the independent t-test or Mann-Whitney U test to identify CAS-specific prognostic biomarkers. Kaplan-Meier and Cox proportional hazards analyses with adjustment for baseline demographic and clinical characteristics were performed to assess the prognostic value of differentially expressed inflammatory proteins in predicting a 2-year MACE in patients with CAS. Results: The mean age of the cohort was 68.8 (SD 10.2) years and 39% were female. The plasma concentrations of two inflammatory proteins were significantly higher in individuals with a 2-year MACE relative to those without a 2-year MACE: IL-6 (5.07 (SD 4.66) vs. 3.36 (SD 4.04) pg/mL, p = 0.03) and CD163 (233.825 (SD 230.306) vs. 159.673 (SD 175.669) pg/mL, p = 0.033). Over a follow-up period of 2 years, individuals with elevated levels of IL-6 were more likely to develop MACE (HR 1.269 (95% CI 1.122-1.639), p = 0.042). Similarly, over a 2-year period, patients with high levels of CD163 were more likely to develop MACE (HR 1.413 (95% CI 1.022-1.954), p = 0.036). Conclusions: The plasma levels of inflammatory proteins IL-6 and CD163 are independently associated with adverse outcomes in individuals with CAS. These CAS-specific prognostic biomarkers may assist in the risk stratification of patients at an elevated risk of a MACE and subsequently guide further vascular evaluation, specialist referrals, and aggressive medical/surgical management, thereby improving outcomes for patients with CAS.


Subject(s)
Biomarkers , Carotid Stenosis , Humans , Female , Carotid Stenosis/blood , Carotid Stenosis/complications , Male , Biomarkers/blood , Aged , Middle Aged , Prospective Studies , Inflammation/blood , Inflammation/complications , Receptors, Cell Surface/blood , Prognosis , Interleukin-6/blood , Proportional Hazards Models , Antigens, CD/blood , Antigens, Differentiation, Myelomonocytic/blood , Kaplan-Meier Estimate , Myocardial Infarction/blood , Myocardial Infarction/complications , Cardiovascular Diseases/blood , Stroke/blood , Stroke/etiology
15.
Microb Pathog ; 193: 106712, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38851360

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) is a major challenge for the global swine industry, causing huge economic losses worldwide. To date, there are no effective measures to prevent and control the spread of PRRS virus (PRRSV). Baicalin (BA) is a natural flavonoid with various pharmacological effects, including antiviral, anti-inflammatory, antioxidant and immunomodulatory. Here, we demonstrate that BA exhibits potent anti-PRRSV activity in vitro, BA concentrations in the range of 5-20 µg/mL significantly inhibited PRRSV infection in a dose-dependent manner and were independent of PRRSV strain. Mechanistically, BA inhibited PRRSV replication by directly interacting with virions, thereby affecting multiple stages of the virus life cycle. Meanwhile, the preventive effect of BA on PRRSV could be realized by inhibiting CD151 and CD163 expression. Furthermore, BA reduced the PRRSV-induced expression of PAMs cytokines (IFN-α, IL-6, IL-8, and TNF-α), suggesting that BA-induced antiviral cytokines may help BA inhibit PRRSV infection. Taken together, BA can be used as an inhibitor of PRRSV infection in vitro, which provides a theoretical basis for the clinical application of BA and the prevention and control of PRRSV infection, which is worthy of further in vivo studies in swine.


Subject(s)
Antiviral Agents , Cytokines , Flavonoids , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Virus Replication , Porcine respiratory and reproductive syndrome virus/drug effects , Animals , Flavonoids/pharmacology , Antiviral Agents/pharmacology , Swine , Virus Replication/drug effects , Cytokines/metabolism , Porcine Reproductive and Respiratory Syndrome/virology , Porcine Reproductive and Respiratory Syndrome/drug therapy , Antigens, CD/metabolism , Receptors, Cell Surface/metabolism , Cell Line , Antigens, Differentiation, Myelomonocytic
16.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891800

ABSTRACT

Sports-related muscle injuries account for 10-55% of all injuries, which is a growing concern, especially given the aging world population. To evaluate the process of skeletal muscle injury and compare it with muscle lesions observed in humans, we developed a novel in vivo model in sheep. In this model, muscle injury was induced by an ultrasound-guided transverse biopsy at the myotendinous junction of the medial gastrocnemius muscle. Twelve male sheep were examined at 3, 7, 14, and 28 days post-injury. Histological, immunofluorescence, and MRI analyses indicate that our sheep model could resemble key human clinicopathological features. Statistically significant differences (p < 0.05) were observed in collagen I, dMHC, α-SMA, and CD68 immunohistochemical detection when comparing injured and healthy muscles. The injured gastrocnemius muscle exhibited elevated levels of type I collagen, infiltration of CD68(+) macrophages, angiogenesis, and the emergence of newly regenerated dMHC(+) myofibers, which persisted for up to 4 weeks post-injury. Similarly, the progression of muscle injury in the sheep model was assessed using advanced clinical 3 T MRI and compared with MRI scans from human patients. The data indicate that the sheep muscle injury model presents features similar to those observed in human skeletal muscle injuries. This makes it a valuable large animal model for studying muscle injuries and developing novel therapeutic strategies.


Subject(s)
Disease Models, Animal , Magnetic Resonance Imaging , Muscle, Skeletal , Animals , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Sheep , Male , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Humans , Collagen Type I/metabolism , Minimally Invasive Surgical Procedures/methods
17.
Sci Rep ; 14(1): 13899, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38886354

ABSTRACT

The current investigation aims to study the embryonic dermis formed in the early stages of development and identify the initial interstitial components of the dermis that serve as biological and structural scaffolds for the development of the dermal tissue. To investigate the dermal structure, the current study used morphological and immunological techniques. TCs identified by TEM. They had a cell body and unique podomeres and podoms. They formed a 3D network spread throughout the dermis. Homocellular contact established between them, as well as heterocellular contacts with other cells. Immunohistochemical techniques using specific markers for TCss CD34, CD117, and VEGF confirmed TC identification. TCs represent the major interstitial component in the dermal tissue. They established a 3D network, enclosing other cells and structures. Expression of VEGF by TC promotes angiogenesis. TCs establish cellular contact with sprouting endothelial cells. At the site of cell junction with TCs, cytoskeletal filaments identified and observed to form the pseudopodium core that projects from endothelial cells. TCs had proteolytic properties that expressed MMP-9, CD68, and CD21. Proteolytic activity aids in the removal of components of the extracellular matrix and the phagocytosis of degraded remnants to create spaces to facilitate the development of new dermal structures. In conclusion, TCs organized the scaffold for the development of future dermal structures, including fibrous components and skin appendages. Studying dermal TCs would be interested in the possibility of developing therapeutic strategies for treating different skin disorders and diseases.


Subject(s)
Dermis , Immunohistochemistry , Telocytes , Telocytes/metabolism , Telocytes/cytology , Dermis/metabolism , Dermis/cytology , Humans , Antigens, CD34/metabolism , Animals , Vascular Endothelial Growth Factor A/metabolism , Antigens, CD/metabolism , Matrix Metalloproteinase 9/metabolism , Endothelial Cells/metabolism , Endothelial Cells/cytology , Antigens, Differentiation, Myelomonocytic/metabolism , CD68 Molecule
18.
Emerg Microbes Infect ; 13(1): 2366359, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38855910

ABSTRACT

Tuberculosis (TB) remains a leading cause of mortality among individuals coinfected with HIV, characterized by progressive pulmonary inflammation. Despite TB's hallmark being focal granulomatous lung lesions, our understanding of the histopathological features and regulation of inflammation in HIV & TB coinfection remains incomplete. In this study, we aimed to elucidate these histopathological features through an immunohistochemistry analysis of HIV & TB co-infected and TB patients, revealing marked differences. Notably, HIV & TB granulomas exhibited aggregation of CD68 + macrophage (Mφ), while TB lesions predominantly featured aggregation of CD20+ B cells, highlighting distinct immune responses in coinfection. Spatial transcriptome profiling further elucidated CD68+ Mφ aggregation in HIV & TB, accompanied by activation of IL6 pathway, potentially exacerbating inflammation. Through multiplex immunostaining, we validated two granuloma types in HIV & TB versus three in TB, distinguished by cell architecture. Remarkably, in the two types of HIV & TB granulomas, CD68 + Mφ highly co-expressed IL6R/pSTAT3, contrasting TB granulomas' high IFNGRA/SOCS3 expression, indicating different signaling pathways at play. Thus, activation of IL6 pathway may intensify inflammation in HIV & TB-lungs, while SOCS3-enriched immune microenvironment suppresses IL6-induced over-inflammation in TB. These findings provide crucial insights into HIV & TB granuloma formation, shedding light on potential therapeutic targets, particularly for granulomatous pulmonary under HIV & TB co-infection. Our study emphasizes the importance of a comprehensive understanding of the immunopathogenesis of HIV & TB coinfection and suggests potential avenues for targeting IL6 signaling with SOCS3 activators or anti-IL6R agents to mitigate lung inflammation in HIV & TB coinfected individuals.


Subject(s)
Coinfection , Granuloma , HIV Infections , Lung , Macrophages , Receptors, Interleukin-6 , STAT3 Transcription Factor , Humans , Coinfection/virology , Coinfection/immunology , Coinfection/microbiology , HIV Infections/complications , HIV Infections/immunology , Macrophages/immunology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Granuloma/immunology , Lung/pathology , Lung/immunology , Receptors, Interleukin-6/metabolism , Receptors, Interleukin-6/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, CD/metabolism , Antigens, CD/genetics , Signal Transduction , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/complications , Male , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/complications , Female , Adult , Interleukin-6/metabolism , Interleukin-6/genetics , CD68 Molecule
19.
Article in Russian | MEDLINE | ID: mdl-38881016

ABSTRACT

BACKGROUND: Contrast enhancement of intracranial aneurysm wall during MRI with targeted visualization of vascular wall correlates with previous aneurysm rupture and, according to some data, may be a predictor of further rupture of unruptured aneurysms. OBJECTIVE: To analyze possible causes of aneurysm contrast enhancement considering morphological data of aneurysm walls. MATERIAL AND METHODS: The study included 44 patients with intracranial aneurysms who underwent preoperative MRI between November 2020 and September 2022. Each aneurysm was assessed regarding contrast enhancement pattern. Microsurgical treatment of aneurysm was accompanied by resection of its wall for subsequent histological and immunohistochemical analysis regarding thrombosis, inflammation and neovascularization. Specimens were subjected to histological and immunochemical analysis. Immunohistochemical analysis was valuable to estimate inflammatory markers CD68 and CD3, as well as neurovascularization marker SD31. RESULTS: Aneurysms with contrast-enhanced walls were characterized by higher number of CD3+, CD68+, CD31+ cells and parietal clots. Intensity of contrast enhancement correlated with aneurysm wall abnormalities. CONCLUSION: Contrast enhancement of aneurysm wall can characterize various morphological abnormalities.


Subject(s)
Intracranial Aneurysm , Magnetic Resonance Imaging , Humans , Intracranial Aneurysm/diagnostic imaging , Intracranial Aneurysm/surgery , Intracranial Aneurysm/pathology , Male , Female , Middle Aged , Magnetic Resonance Imaging/methods , Neuroimaging/methods , Antigens, Differentiation, Myelomonocytic/analysis , Antigens, Differentiation, Myelomonocytic/metabolism , Adult , Contrast Media , Antigens, CD/analysis , Antigens, CD/metabolism , Aged , Aneurysm, Ruptured/diagnostic imaging , Aneurysm, Ruptured/surgery , Aneurysm, Ruptured/pathology , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , CD3 Complex/analysis , CD3 Complex/metabolism , CD68 Molecule
20.
Nat Commun ; 15(1): 5180, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890323

ABSTRACT

Siglec-6 is a lectin receptor with restricted expression in the placenta, mast cells and memory B-cells. Although Siglec-6 is expressed in patients with chronic lymphocytic leukemia (CLL), its pathophysiological role has not been elucidated. We describe here a role for Siglec-6 in migration and adhesion of CLL B cells to CLL- bone marrow stromal cells (BMSCs) in vitro and compromised migration to bone marrow and spleen in vivo. Mass spectrometry analysis revealed interaction of Siglec-6 with DOCK8, a guanine nucleotide exchange factor. Stimulation of MEC1-002 CLL cells with a Siglec-6 ligand, sTn, results in Cdc42 activation, WASP protein recruitment and F-actin polymerization, which are all associated with cell migration. Therapeutically, a Siglec-6/CD3-bispecific T-cell-recruiting antibody (T-biAb) improves overall survival in an immunocompetent mouse model and eliminates CLL cells in a patient derived xenograft model. Our findings thus reveal a migratory role for Siglec-6 in CLL, which can be therapeutically targeted using a Siglec-6 specific T-biAb.


Subject(s)
Cell Adhesion , Cell Movement , Lectins , Leukemia, Lymphocytic, Chronic, B-Cell , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Humans , Animals , Lectins/metabolism , Mice , Antigens, CD/metabolism , Antigens, CD/genetics , Female , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Cell Line, Tumor , Mesenchymal Stem Cells/metabolism , Male , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL