Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
1.
Gut ; 73(9): 1543-1553, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-38724220

ABSTRACT

OBJECTIVE: Previous studies indicate that eosinophils are recruited into the allograft following orthotopic liver transplantation and protect from ischaemia reperfusion (IR) injury. In the current studies, we aim to explore whether their protective function could outlast during liver repair. DESIGN: Eosinophil-deficient mice and adoptive transfer of bone marrow-derived eosinophils (bmEos) were employed to investigate the effects of eosinophils on tissue repair and regeneration after hepatic IR injury. Aside from exogenous cytokine or neutralising antibody treatments, mechanistic studies made use of a panel of mouse models of eosinophil-specific IL-4/IL-13-deletion, cell-specific IL-4rα-deletion in liver macrophages and hepatocytes and macrophage-specific deletion of heparin-binding epidermal growth factor-like growth factor (hb-egf). RESULT: We observed that eosinophils persisted over a week following hepatic IR injury. Their peak accumulation coincided with that of hepatocyte proliferation. Functional studies showed that eosinophil deficiency was associated with a dramatic delay in liver repair, which was normalised by the adoptive transfer of bmEos. Mechanistic studies demonstrated that eosinophil-derived IL-4, but not IL-13, was critically involved in the reparative function of these cells. The data further revealed a selective role of macrophage-dependent IL-4 signalling in liver regeneration. Eosinophil-derived IL-4 stimulated macrophages to produce HB-EGF. Moreover, macrophage-specific hb-egf deletion impaired hepatocyte regeneration after IR injury. CONCLUSION: Together, these studies uncovered an indispensable role of eosinophils in liver repair after acute injury and identified a novel crosstalk between eosinophils and macrophages through the IL-4/HB-EGF axis.


Subject(s)
Eosinophils , Heparin-binding EGF-like Growth Factor , Interleukin-4 , Liver Regeneration , Macrophages , Reperfusion Injury , Animals , Heparin-binding EGF-like Growth Factor/metabolism , Heparin-binding EGF-like Growth Factor/genetics , Liver Regeneration/physiology , Reperfusion Injury/metabolism , Interleukin-4/metabolism , Mice , Eosinophils/metabolism , Macrophages/metabolism , Liver/pathology , Liver/metabolism , Liver/blood supply , Hepatocytes/metabolism , Interleukin-13/metabolism , Adoptive Transfer , Mice, Inbred C57BL
2.
FEBS J ; 291(10): 2094-2097, 2024 May.
Article in English | MEDLINE | ID: mdl-38680125

ABSTRACT

Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is a transmembrane protein that, when cleaved by metalloproteases through a process called ectodomain shedding, binds to the EGF receptor (EGFR), activating downstream signaling. The HB-EGF/EGFR pathway is crucial in development and is involved in numerous pathophysiological processes. In this issue of The FEBS Journal, Sireci et al. reveal a previously unexplored function of the HB-EGF/EGFR pathway in promoting neuronal progenitor proliferation and sensory neuron regeneration in the zebrafish olfactory epithelium in response to injury.


Subject(s)
ErbB Receptors , Heparin-binding EGF-like Growth Factor , Signal Transduction , Zebrafish , Animals , Humans , Cell Proliferation , ErbB Receptors/metabolism , ErbB Receptors/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Heparin-binding EGF-like Growth Factor/genetics , Nerve Regeneration , Neurons/metabolism , Neurons/pathology , Olfactory Mucosa/metabolism , Zebrafish/metabolism
3.
Nat Immunol ; 25(3): 432-447, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38409259

ABSTRACT

Central nervous system (CNS)-resident cells such as microglia, oligodendrocytes and astrocytes are gaining increasing attention in respect to their contribution to CNS pathologies including multiple sclerosis (MS). Several studies have demonstrated the involvement of pro-inflammatory glial subsets in the pathogenesis and propagation of inflammatory events in MS and its animal models. However, it has only recently become clear that the underlying heterogeneity of astrocytes and microglia can not only drive inflammation, but also lead to its resolution through direct and indirect mechanisms. Failure of these tissue-protective mechanisms may potentiate disease and increase the risk of conversion to progressive stages of MS, for which currently available therapies are limited. Using proteomic analyses of cerebrospinal fluid specimens from patients with MS in combination with experimental studies, we here identify Heparin-binding EGF-like growth factor (HB-EGF) as a central mediator of tissue-protective and anti-inflammatory effects important for the recovery from acute inflammatory lesions in CNS autoimmunity. Hypoxic conditions drive the rapid upregulation of HB-EGF by astrocytes during early CNS inflammation, while pro-inflammatory conditions suppress trophic HB-EGF signaling through epigenetic modifications. Finally, we demonstrate both anti-inflammatory and tissue-protective effects of HB-EGF in a broad variety of cell types in vitro and use intranasal administration of HB-EGF in acute and post-acute stages of autoimmune neuroinflammation to attenuate disease in a preclinical mouse model of MS. Altogether, we identify astrocyte-derived HB-EGF and its epigenetic regulation as a modulator of autoimmune CNS inflammation and potential therapeutic target in MS.


Subject(s)
Astrocytes , Multiple Sclerosis , Animals , Humans , Mice , Anti-Inflammatory Agents , Disease Models, Animal , Epigenesis, Genetic , Heparin-binding EGF-like Growth Factor/genetics , Inflammation , Proteomics
4.
Nat Commun ; 14(1): 4857, 2023 08 11.
Article in English | MEDLINE | ID: mdl-37567873

ABSTRACT

Unlike adult mammals, zebrafish regenerate spinal cord tissue and recover locomotor ability after a paralyzing injury. Here, we find that ependymal cells in zebrafish spinal cords produce the neurogenic factor Hb-egfa upon transection injury. Animals with hb-egfa mutations display defective swim capacity, axon crossing, and tissue bridging after spinal cord transection, associated with disrupted indicators of neuron production. Local recombinant human HB-EGF delivery alters ependymal cell cycling and tissue bridging, enhancing functional regeneration. Epigenetic profiling reveals a tissue regeneration enhancer element (TREE) linked to hb-egfa that directs gene expression in spinal cord injuries. Systemically delivered recombinant AAVs containing this zebrafish TREE target gene expression to crush injuries of neonatal, but not adult, murine spinal cords. Moreover, enhancer-based HB-EGF delivery by AAV administration improves axon densities after crush injury in neonatal cords. Our results identify Hb-egf as a neurogenic factor necessary for innate spinal cord regeneration and suggest strategies to improve spinal cord repair in mammals.


Subject(s)
Spinal Cord Injuries , Spinal Cord Regeneration , Animals , Humans , Mice , Axons/metabolism , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Mammals , Nerve Regeneration/genetics , Spinal Cord/metabolism , Spinal Cord Injuries/genetics , Spinal Cord Injuries/therapy , Spinal Cord Injuries/metabolism , Spinal Cord Regeneration/physiology , Zebrafish/genetics
5.
Proc Natl Acad Sci U S A ; 120(20): e2302937120, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37155852

ABSTRACT

Implantation is the first direct encounter between the embryo and uterus during pregnancy, and Hbegf is the earliest known molecular signaling for embryo-uterine crosstalk during implantation. The downstream effectors of heparin-binding EGF (HB-EGF) in implantation remain elusive due to the complexity of EGF receptor family. This study shows that the formation of implantation chamber (crypt) triggered by HB-EGF is disrupted by uterine deletion of Vangl2, a key planar cell polarity component (PCP). We found that HB-EGF binds to ERBB2 and ERBB3 to recruit VANGL2 for tyrosine phosphorylation. Using in vivo models, we show that uterine VAGL2 tyrosine phosphorylation is suppressed in Erbb2/Erbb3 double conditional knockout mice. In this context, severe implantation defects in these mice lend support to the critical role of HB-EGF-ERBB2/3-VANGL2 in establishing a two-way dialogue between the blastocyst and uterus. In addition, the result addresses an outstanding question how VANGL2 is activated during implantation. Taken together, these observations reveal that HB-EGF regulates the implantation process by influencing uterine epithelial cell polarity comprising VANGL2.


Subject(s)
Cell Polarity , Embryo Implantation , Animals , Female , Mice , Pregnancy , Cell Polarity/physiology , Embryo Implantation/physiology , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Mice, Knockout , Signal Transduction , Tyrosine
6.
JCI Insight ; 7(18)2022 09 22.
Article in English | MEDLINE | ID: mdl-35998057

ABSTRACT

Macrophages in the tumor microenvironment have a substantial impact on tumor progression. Depending on the signaling environment in the tumor, macrophages can either support or constrain tumor progression. It is therefore of therapeutic interest to identify the tumor-derived factors that control macrophage education. With this aim, we correlated the expression of A Disintegrin and Metalloproteinase (ADAM) proteases, which are key mediators of cell-cell signaling, to the expression of protumorigenic macrophage markers in human cancer cohorts. We identified ADAM17, a sheddase upregulated in many cancer types, as a protein of interest. Depletion of ADAM17 in cancer cell lines reduced the expression of several protumorigenic markers in neighboring macrophages in vitro as well as in mouse models. Moreover, ADAM17-/- educated macrophages demonstrated a reduced ability to induce cancer cell invasion. Using mass spectrometry-based proteomics and ELISA, we identified heparin-binding EGF (HB-EGF) and amphiregulin, shed by ADAM17 in the cancer cells, as the implicated molecular mediators of macrophage education. Additionally, RNA-Seq and ELISA experiments revealed that ADAM17-dependent HB-EGF ligand release induced the expression and secretion of CXCL chemokines in macrophages, which in turn stimulated cancer cell invasion. In conclusion, we provide evidence that ADAM17 mediates a paracrine EGFR-ligand-chemokine feedback loop, whereby cancer cells hijack macrophages to promote tumor progression.


Subject(s)
ADAM17 Protein , Disintegrins , Macrophages , Neoplasm Invasiveness , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Amphiregulin , Animals , Epidermal Growth Factor , ErbB Receptors/metabolism , Heparin , Heparin-binding EGF-like Growth Factor/genetics , Humans , Ligands , Macrophages/metabolism , Mice , Tumor Microenvironment
7.
Cell Cycle ; 21(23): 2517-2532, 2022 12.
Article in English | MEDLINE | ID: mdl-35899982

ABSTRACT

Preeclampsia (PE) is a pregnancy-associated disease that may cause maternal and fetal morbidity and mortality. The dysregulation of microRNAs (miRNAs) and their potential functions has been an important direction for elucidating the mechanism of preeclampsia in recent years. The present study investigated whether miR-4443 was significantly increased in the placentas of severe preeclamptic patients, and the upregulation of miR-4443 inhibited the migration and invasion of HTR-8/SVneo cells according to transwell assays. Matrix metallopeptidase 2 (MMP2), which is involved in the degradation of extracellular matrix (ECM) components and harbors a miR-4443-binding site within its 3'-UTR as confirmed by a luciferase reporter assay, was identified to be directly inhibited by miR-4443. Moreover, siRNA targeting MMP2 imitated the effects of overexpressed miR-4443 on HTR-8/SVneo cell invasion and migration, whereas rescue experiments showed that MMP2 reversed this inhibitory function of miR-4443. Heparin-binding EGF-like growth factor (HB-EGF), as the downstream gene of MMP2, plays an important role in trophoblast invasion, and we confirmed that the expression of HB-EGF/EGFR pathway-related biomolecules was consistent with MMP2 influenced by upregulating and downregulating miR-4443 and that activated EGFR further transmitted intracellular downstream signaling via the MAPK pathway according to western blot assay. In conclusion, we demonstrated that miR-4443 suppresses the migration and invasion of trophoblasts, and its inhibitory effects are at least partially mediated by the suppression of MMP2. This inhibition might further affect the progression of preeclampsia through the HB-EGF/EGFR pathway, thus providing a new clue on the role of miR-4443 in the pathogenesis of preeclampsia.


Subject(s)
MicroRNAs , Pre-Eclampsia , Pregnancy , Female , Humans , Trophoblasts/metabolism , Pre-Eclampsia/pathology , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Cell Movement/genetics , Cell Line , MicroRNAs/genetics , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Cell Proliferation
8.
STAR Protoc ; 3(2): 101411, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35620071

ABSTRACT

Intestinal cells marked by Lgr5 function as tissue-resident stem cells that sustain the homeostatic replenishment of the epithelium. By incorporating a diphtheria toxin receptor (DTR) cassette linked to the Lgr5 coding region, native Lgr5-expressing cells are susceptible to ablation upon DT administration in vivo. A similar strategy can be used for Lgr5-expressing cells within organoids established from DTR models. Together, these in vivo and in vitro approaches will facilitate dissection of the roles of Lgr5-expressing cells residing in different tissue compartments. For complete details on the use and execution of this protocol, please refer to Tan et al. (2021).


Subject(s)
Organoids , Receptors, G-Protein-Coupled , Animals , Heparin-binding EGF-like Growth Factor/genetics , Intestines , Mice , Receptors, G-Protein-Coupled/genetics , Stem Cells
9.
World J Gastroenterol ; 28(17): 1781-1797, 2022 May 07.
Article in English | MEDLINE | ID: mdl-35633908

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is an extremely malignant tumor with a high mortality rate. Little is known about the mechanism by which forkhead Box q1 (FOXQ1) causes CRC invasion and metastasis through the epidermal growth factor receptor (EGFR) pathway. AIM: To illuminate the mechanism by which FOXQ1 promotes the invasion and metastasis of CRC by activating the heparin binding epidermal growth factor (HB-EGF)/EGFR pathway. METHODS: We investigated the differential expression and prognosis of FOXQ1 and HB-EGF in CRC using the Gene Expression Profiling Interactive Analysis (GEPIA) website (http://gepia.cancer-pku.cn/index.html). Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting were used to detect the expression of FOXQ1 and HB-EGF in cell lines and tissues, and we constructed a stable low-expressing FOXQ1 cell line and verified it with the above method. The expression changes of membrane-bound HB-EGF (proHB-EGF) and soluble HB-EGF (sHB-EGF) in the low-expressing FOXQ1 cell line were detected by flow cytometry and ELISA. Western blotting was used to detect changes in the expression levels of HB-EGF and EGFR pathway-related downstream genes when exogenous recombinant human HB-EGF was added to FOXQ1 knockdown cells. Proliferation experiments, transwell migration experiments, and scratch experiments were carried out to determine the mechanism by which FOXQ1 activates the EGFR signaling pathway through HB-EGF, and then to evaluate the clinical relevance of FOXQ1 and HB-EGF. RESULTS: GEPIA showed that the expression of FOXQ1 in CRC tissues was relatively high and was related to a lower overall survival rate. PCR array results showed that FOXQ1 is related to the HB-EGF and EGFR pathways. Knockdown of FOXQ1 suppressed the expression of HB-EGF, and led to a decrease in EGFR and its downstream genes AKT, RAF, KRAS expression levels. After knockdown of FOXQ1 in CRC cell lines, cell proliferation, migration and invasion were attenuated. Adding HB-EGF restored the migration and invasion ability of CRC, but not the cell proliferation ability. Kaplan-Meier survival analysis results showed that the combination of FOXQ1 and HB-EGF may serve to predict CRC survival. CONCLUSION: Based on these collective data, we propose that FOXQ1 promotes the invasion and metastasis of CRC via the HB-EGF/EGFR pathway.


Subject(s)
Colorectal Neoplasms , Epidermal Growth Factor , Forkhead Transcription Factors , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Neoplasm Invasiveness , Neoplasm Metastasis
10.
BMC Cancer ; 22(1): 254, 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35264144

ABSTRACT

BACKGROUND: The standard treatment for patients with advanced HER2-positive gastric cancer is a combination of the antibody trastuzumab and platin-fluoropyrimidine chemotherapy. As some patients do not respond to trastuzumab therapy or develop resistance during treatment, the search for alternative treatment options and biomarkers to predict therapy response is the focus of research. We compared the efficacy of trastuzumab and other HER-targeting drugs such as cetuximab and afatinib. We also hypothesized that treatment-dependent regulation of a gene indicates its importance in response and that it can therefore be used as a biomarker for patient stratification. METHODS: A selection of gastric cancer cell lines (Hs746T, MKN1, MKN7 and NCI-N87) was treated with EGF, cetuximab, trastuzumab or afatinib for a period of 4 or 24 h. The effects of treatment on gene expression were measured by RNA sequencing and the resulting biomarker candidates were tested in an available cohort of gastric cancer patients from the VARIANZ trial or functionally analyzed in vitro. RESULTS: After treatment of the cell lines with afatinib, the highest number of regulated genes was observed, followed by cetuximab and trastuzumab. Although trastuzumab showed only relatively small effects on gene expression, BMF, HAS2 and SHB could be identified as candidate biomarkers for response to trastuzumab. Subsequent studies confirmed HAS2 and SHB as potential predictive markers for response to trastuzumab therapy in clinical samples from the VARIANZ trial. AREG, EREG and HBEGF were identified as candidate biomarkers for treatment with afatinib and cetuximab. Functional analysis confirmed that HBEGF is a resistance factor for cetuximab. CONCLUSION: By confirming HAS2, SHB and HBEGF as biomarkers for anti-HER therapies, we provide evidence that the regulation of gene expression after treatment can be used for biomarker discovery. TRIAL REGISTRATION: Clinical specimens of the VARIANZ study (NCT02305043) were used to test biomarker candidates.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Heparin-binding EGF-like Growth Factor/genetics , Hyaluronan Synthases/genetics , Proto-Oncogene Proteins/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Afatinib/pharmacology , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cetuximab/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression/drug effects , Humans , Receptor, ErbB-2/drug effects , Trastuzumab/pharmacology
11.
Front Cell Infect Microbiol ; 12: 823714, 2022.
Article in English | MEDLINE | ID: mdl-35281434

ABSTRACT

Otitis media (OM), the most common childhood illness, can be caused by bacterial and/or viral infection. Hyperplasia of the middle ear (ME) mucosa is an important component of OM that contributes to its deleterious sequelae. Our previous research revealed that ME mucosal hyperplasia in bacterially induced OM was associated with expression of the heparin-binding epidermal growth factor (HB-EGF) gene, and that HB-EGF induced the proliferation of ME mucosal explants in culture. We used single-cell RNA-Seq to identify ME cells that express Hbegf and related genes involved in mediating responses to this factor. To determine the degree to which a viral infection might induce mucosal hyperplasia, and to assess the role of HB-EGF in hyperplasia in vivo, we used, Poly(I:C) to simulate a ME viral infection, Western blotting to confirm ME protein expression, and a specific inhibitor to block the effects of HB-EGF during OM. Genes for HB-EGF and its receptor were expressed in the ME primarily by epithelial, stromal and endothelial cells. Poly(I:C) induced prominent ME mucosal hyperplasia, peaking two days after ME injection. Immunostaining revealed that cleavage of proHB-EGF into its soluble form (sHB-EGF) was strongly induced in response to Poly(I:C). Inhibition of the sHB-EGF receptor dramatically reduced the hyperplastic response of the mucosa. The results demonstrate that a synthetic analog of viral double-stranded RNA interaction can induce OM including a strong proliferative response of the ME mucosa, independent of bacteria. They also indicate that HB-EGF is the dominant growth factor responsible for ME mucosal hyperplasia in vivo.


Subject(s)
Epidermal Growth Factor , Heparin-binding EGF-like Growth Factor , Otitis Media , Virus Diseases , Animals , Endothelial Cells/metabolism , Epidermal Growth Factor/metabolism , Female , Heparin-binding EGF-like Growth Factor/adverse effects , Heparin-binding EGF-like Growth Factor/genetics , Humans , Hyperplasia , Male , Mice, Inbred C57BL , Rats, Sprague-Dawley , Virus Diseases/metabolism
12.
Int J Biol Sci ; 18(5): 2047-2059, 2022.
Article in English | MEDLINE | ID: mdl-35342363

ABSTRACT

Polycystic ovarian syndrome (PCOS) is one of the most prevalent endocrinopathies and the leading cause of anovulatory infertility, but its pathogenesis remains elusive. Although HB-EGF is involved in ovarian cancer progression, there is still no clarity about its relevance with PCOS. The present study exhibited that abundant HB-EGF was noted in follicular fluid from PCOS women, where it might induce the granulosa cells (GCs) production of more estrogen via the elevation of CYP19A1 expression after binding to EGFR. Furthermore, HB-EGF transduced intracellular downstream cAMP-PKA signaling to promote the phosphorylation of JNK and ERK whose blockage impeded the induction of HB-EGF on estrogen secretion. Meanwhile, HB-EGF enhanced the accumulation of intracellular Ca2+ whose chelation by BAPTA-AM abrogated the stimulation of HB-EGF on FOXO1 along with an obvious diminishment for estrogen production. cAMP-PKA-JNK/ERK-Ca2+ pathway played an important role in the crosstalk between HB-EGF and FOXO1. Treatment of GCs with HB-EGF resulted in mitochondrial dysfunction as evinced by the reduction of ATP content, mtDNA copy number and mitochondrial membrane potential. Additionally, HB-EGF facilitated the opening of mitochondrial permeability transition pore via targeting BAX and raised the release of cytochrome C from mitochondria into the cytosol to trigger the apoptosis of GCs, but this effectiveness was counteracted by estrogen receptor antagonist. Collectively, HB-EGF might induce mitochondrial dysfunction and GCs apoptosis through advancing estrogen hypersecretion dependent on cAMP-PKA-JNK/ERK-Ca2+-FOXO1 pathway and act as a promising therapeutic target for PCOS.


Subject(s)
Polycystic Ovary Syndrome , Estrogens/metabolism , Estrogens/pharmacology , Female , Forkhead Box Protein O1/metabolism , Granulosa Cells/metabolism , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Heparin-binding EGF-like Growth Factor/pharmacology , Humans , Mitochondria/metabolism , Polycystic Ovary Syndrome/metabolism
13.
Biochem Biophys Res Commun ; 599: 113-119, 2022 04 09.
Article in English | MEDLINE | ID: mdl-35180470

ABSTRACT

Pulmonary fibrosis (PF) is a progressive fibrotic disease with poor prognosis and suboptimal therapeutic options. Although macrophages have been implicated in PF, the role of macrophage subsets, particularly interstitial macrophages (IMs), remains unknown. We performed a time-series single-cell RNA sequencing analysis of the silica-induced mouse PF model. Among the macrophage subsets in fibrotic lungs, Lyve1lo MHC IIhi IMs increased with fibrosis, and highly expressed profibrotic genes. Additionally, we identified C1q as an IM-specific marker. Experiments with C1q-diphtheria toxin receptor-GFP knock-in (C1qKI) mice revealed that IMs are distributed around fibrotic nodules. Depletion of C1q+ IMs in C1qKI mice decreased activated fibroblasts and epithelial cells; however, bodyweight loss and neutrophil infiltration were exacerbated in silica-induced PF. Collectively, these results suggest that IMs have profibrotic and anti-inflammatory properties and that the selective inhibition of the profibrotic function of IMs without compromising their anti-inflammatory effects is a potential novel therapeutic strategy for PF.


Subject(s)
Complement C1q/metabolism , Macrophages/pathology , Pulmonary Fibrosis/pathology , Animals , Biomarkers/metabolism , Complement C1q/genetics , Disease Models, Animal , Gene Expression , Heparin-binding EGF-like Growth Factor/genetics , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Silicon Dioxide/toxicity
14.
J Neuroendocrinol ; 33(12): e13057, 2021 12.
Article in English | MEDLINE | ID: mdl-34748241

ABSTRACT

Vasopressin-synthesizing neurons are located in several brain regions, including the hypothalamic paraventricular nucleus (PVN), supraoptic nucleus (SON) and suprachiasmatic nucleus (SCN). Vasopressin has been shown to have various functions in the brain, including social recognition memory, stress responses, emotional behaviors and circadian rhythms. The precise physiological functions of vasopressin-synthesizing neurons in specific brain regions remain to be clarified. Conditional ablation of local vasopressin-synthesizing neurons may be a useful tool for investigation of the functions of vasopressin neurons in the regions. In the present study, we characterized a transgenic rat line that expresses a mutated human diphtheria toxin receptor under control of the vasopressin gene promoter. Under a condition of salt loading, which activates the vasopressin gene in the hypothalamic PVN and SON, transgenic rats were i.c.v. injected with diphtheria toxin. Intracerebroventricular administration of diphtheria toxin after salt loading depleted vasopressin-immunoreactive cells in the hypothalamic PVN and SON, but not in the SCN. The number of oxytocin-immunoreactive cells in the hypothalamus was not significantly changed. The rats that received i.c.v. diphtheria toxin after salt loading showed polydipsia and polyuria, which were rescued by peripheral administration of 1-deamino-8-d-arginine vasopressin via an osmotic mini-pump. Intrahypothalamic administration of diphtheria toxin in transgenic rats under a normal hydration condition reduced the number of vasopressin-immunoreactive neurons, but not the number of oxytocin-immunoreactive neurons. The transgenic rat model can be used for selective ablation of vasopressin-synthesizing neurons and may be useful for clarifying roles of vasopressin neurons at least in the hypothalamic PVN and SON in the rat.


Subject(s)
Gene Transfer Techniques , Genes, Transgenic, Suicide , Neurons/metabolism , Vasopressins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Diphtheria Toxin/pharmacology , Gene Deletion , Genes, Transgenic, Suicide/drug effects , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Male , Neurons/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Inbred Lew , Rats, Transgenic , Supraoptic Nucleus/drug effects , Supraoptic Nucleus/metabolism , Vasopressins/genetics
15.
Anticancer Res ; 41(9): 4249-4258, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34475044

ABSTRACT

BACKGROUND/AIM: Recent studies have indicated the clinical significance of tumor-associated macrophages (TAMs) in breast cancer; however, the detailed mechanisms of cell-cell interactions between TAMs and cancer cells remain unclear. MATERIALS AND METHODS: In vitro cell culture studies using human monocyte-derived macrophages and breast cancer cell lines were performed to test which cytokines would be involved in cell-cell interactions between cancer cells and macrophages. In addition, studies using human resected samples and animal breast cancer models were performed to examine the significance of TAMs in cancer development. RESULTS: Osteopontin, HB-EGF, and IL-6 were suggested to be macrophage-derived growth factors for breast cancer cells. FROUNT inhibitor significantly blocked TAM infiltration and subcutaneous tumor growth in an E0771 mouse breast cancer model. CONCLUSION: TAMs express growth factors, such as osteopontin, for cancer cells, and targeting of TAM infiltration might be a promising approach for anti-breast cancer therapy.


Subject(s)
Breast Neoplasms/pathology , Heparin-binding EGF-like Growth Factor/genetics , Interleukin-6/genetics , Macrophages/cytology , Osteopontin/genetics , Tumor-Associated Macrophages/cytology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Communication , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cells, Cultured , Coculture Techniques , Female , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Interleukin-6/metabolism , MCF-7 Cells , Macrophages/metabolism , Mice , Neoplasm Transplantation , Osteopontin/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
16.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34502404

ABSTRACT

Rodent models of Parkinson's disease are based on transgenic expression of mutant synuclein, deletion of PD genes, injections of MPTP or rotenone, or seeding of synuclein fibrils. The models show histopathologic features of PD such as Lewi bodies but mostly only subtle in vivo manifestations or systemic toxicity. The models only partly mimic a predominant loss of dopaminergic neurons in the substantia nigra. We therefore generated mice that express the transgenic diphtheria toxin receptor (DTR) specifically in DA neurons by crossing DAT-Cre mice with Rosa26 loxP-STOP-loxP DTR mice. After defining a well-tolerated DTx dose, DAT-DTR and DTR-flfl controls were subjected to non-toxic DTx treatment (5 × 100 pg/g) and subsequent histology and behavioral tests. DAT protein levels were reduced in the midbrain, and tyrosine hydroxylase-positive neurons were reduced in the substantia nigra, whereas the pan-neuronal marker NeuN was not affected. Despite the promising histologic results, there was no difference in motor function tests or open field behavior. These are tests in which double mutant Pink1-/-SNCAA53T Parkinson mice show behavioral abnormalities. Higher doses of DTx were toxic in both groups. The data suggest that DTx treatment in mice with Cre/loxP-driven DAT-DTR expression leads to partial ablation of DA-neurons but without PD-reminiscent behavioral correlates.


Subject(s)
Heparin-binding EGF-like Growth Factor/genetics , Parkinson Disease/physiopathology , Animals , Brain/pathology , Corpus Striatum/metabolism , Diphtheria Toxin/metabolism , Diphtheria Toxin/pharmacology , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/pathology , Female , Heparin-binding EGF-like Growth Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Parkinson Disease/metabolism , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/metabolism
17.
J Mol Biol ; 433(21): 167240, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34508725

ABSTRACT

Receptor tyrosine kinases (RTK) bind growth factors and are critical for cell proliferation and differentiation. Their dysregulation leads to a loss of growth control, often resulting in cancer. Epidermal growth factor receptor (EGFR) is the prototypic RTK and can bind several ligands exhibiting distinct mitogenic potentials. Whereas the phosphorylation on individual EGFR sites and their roles for downstream signaling have been extensively studied, less is known about ligand-specific ubiquitination events on EGFR, which are crucial for signal attenuation and termination. We used a proteomics-based workflow for absolute quantitation combined with mathematical modeling to unveil potentially decisive ubiquitination events on EGFR from the first 30 seconds to 15 minutes of stimulation. Four ligands were used for stimulation: epidermal growth factor (EGF), heparin-binding-EGF like growth factor, transforming growth factor-α and epiregulin. Whereas only little differences in the order of individual ubiquitination sites were observed, the overall amount of modified receptor differed depending on the used ligand, indicating that absolute magnitude of EGFR ubiquitination, and not distinctly regulated ubiquitination sites, is a major determinant for signal attenuation and the subsequent cellular outcomes.


Subject(s)
Epidermal Growth Factor/metabolism , Epiregulin/metabolism , Heparin-binding EGF-like Growth Factor/metabolism , Signal Transduction/genetics , Transforming Growth Factor alpha/metabolism , Amino Acid Sequence , Cell Line, Tumor , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/genetics , Epiregulin/chemistry , Epiregulin/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , ErbB Receptors/chemistry , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression , Heparin-binding EGF-like Growth Factor/chemistry , Heparin-binding EGF-like Growth Factor/genetics , Humans , Ligands , Models, Molecular , Mutation , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational , Proteomics , Transforming Growth Factor alpha/chemistry , Transforming Growth Factor alpha/genetics , Ubiquitination
18.
Mol Cell Endocrinol ; 536: 111418, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34339824

ABSTRACT

Our recent findings indicate that an acute depletion of monocytes has no sustained effects on ovarian follicle health. Here, we utilised a Cx3cr1-Dtr transgenic Wistar rat model to transiently deplete monocytes and investigated the impact of an acute immune challenge by lipopolysaccharide (LPS) on ovarian follicle health and ovulatory capacity relative to wt once the monocytes had repopulated. Monocyte depletion and repopulation exacerbated the effects of LPS in several domains. As such, monocyte perturbation decreased the numbers of secondary follicles in those challenged with LPS. Monocyte perturbation was also associated with reduced antral follicle numbers and circulating luteinising hormone (LH) levels, as well as potential changes in ovarian sensitivity to LH, exacerbated by LPS. These data suggest that monocyte depletion and repopulation induce a transient suppression of ovulatory capacity in response to a subsequent immune challenge, but this is likely to be restored once the pro-inflammatory environment is resolved.


Subject(s)
CX3C Chemokine Receptor 1/genetics , Heparin-binding EGF-like Growth Factor/genetics , Lipopolysaccharides/adverse effects , Ovarian Follicle/metabolism , Animals , Female , Leukocytes, Mononuclear , Lipopolysaccharides/immunology , Luteinizing Hormone/blood , Luteinizing Hormone/pharmacology , Ovarian Follicle/drug effects , Ovarian Follicle/immunology , Promoter Regions, Genetic , Rats , Rats, Transgenic , Rats, Wistar
19.
Front Immunol ; 12: 688347, 2021.
Article in English | MEDLINE | ID: mdl-34194439

ABSTRACT

Mast cells (MCs), strategically localized at mucosal surfaces, provide first-line defense against pathogens and shape innate and adaptive immune responses. Recent studies have shown that MCs are involved in pathogenic responses to several viruses including herpes simplex viruses, dengue virus, vaccinia virus and influenza virus. However, the underlying mechanisms of MCs in the activation of CD8+ T cells during viral infections are not fully understood. Therefore, we investigate the role of MCs in the development of virus-specific CD8+ T cell responses using the well-characterized murine lymphocytic choriomeningitis virus (LCMV) model and the transgenic MasTRECK mice that contain the human diphtheria toxin receptor as an inducible MC-deficient model. Here, we report that MCs are essential for the activation and expansion of virus-specific CD8+ T cells. After MC depletion and subsequent intradermal LCMV infection, the CD8 + T cell effector phenotype and antiviral cytokine production were impaired at the peak of infection (day 8 p.i.). Importantly, MC-deficient mice were unable to control the infection and exhibited significantly higher viral loads in the spleen and in the ear draining lymph nodes compared to that of wild type control mice. In the absence of MCs, dendritic cell (DC) activation was impaired upon LCMV infection. In addition, type-I interferon (IFN) levels in the serum and in the spleen of MC-deficient mice were reduced during the first days of infection. Interestingly, depletion of MCs after intradermal LCMV infection did not impair virus-specific CD8+ T cell expansion, activation or antiviral cytokine production. In summary, our results indicate that MCs play a pivotal role in the activation and antiviral functions of CD8+ T cells through proper DC activation. A better understanding of the impact of MCs on CD8+ T cell responses is mandatory to improve antiviral immune responses.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lymphocyte Activation , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Mast Cells/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Communication , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/metabolism , Dendritic Cells/virology , Disease Models, Animal , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Host-Pathogen Interactions , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/pathogenicity , Mast Cells/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Signal Transduction
20.
Cell Death Dis ; 12(7): 646, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34168124

ABSTRACT

Although macrophages are recognized as important players in the pathogenesis of chronic liver diseases, their roles in cholestatic liver fibrosis remain incompletely understood. We previously reported that long noncoding RNA-H19 (lncRNA-H19) contributes to cholangiocyte proliferation and cholestatic liver fibrosis of biliary atresia (BA). We here show that monocyte/macrophage CD11B mRNA levels are increased significantly in livers of BA patients and positively correlated with the progression of liver inflammation and fibrosis. The macrophages increasingly infiltrate and accumulate in the fibrotic niche and peribiliary areas in livers of BA patients. Selective depletion of macrophages using the transgenic CD11b-diphtheria toxin receptor (CD11b-DTR) mice halts bile duct ligation (BDL)-induced progression of liver damage and fibrosis. Meanwhile, macrophage depletion significantly reduces the BDL-induced hepatic lncRNA-H19. Overexpression of H19 in livers using adeno-associated virus serotype 9 (AAV9) counteracts the effects of macrophage depletion on liver fibrosis and cholangiocyte proliferation. Additionally, both H19 knockout (H19-/-) and conditional deletion of H19 in macrophage (H19ΔCD11B) significantly depress the macrophage polarization and recruitment. lncRNA-H19 overexpressed in THP-1 macrophages enhance expression of Rho-GTPase CDC42 and RhoA. In conclusions, selectively depletion of macrophages suppresses cholestatic liver injuries and fibrosis via the lncRNA-H19 and represents a potential therapeutic strategy for rapid liver fibrosis in BA patients.


Subject(s)
Liver Cirrhosis, Biliary/prevention & control , Liver Cirrhosis, Experimental/prevention & control , Liver/metabolism , Macrophage Activation , Macrophages/metabolism , RNA, Long Noncoding/metabolism , Animals , CD11b Antigen/genetics , CD11b Antigen/metabolism , Case-Control Studies , Cell Proliferation , Cholestasis/complications , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Liver/pathology , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis, Biliary/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , RNA, Long Noncoding/genetics , THP-1 Cells , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL