Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Genesis ; 60(3): e23470, 2022 03.
Article in English | MEDLINE | ID: mdl-35258166

ABSTRACT

The loss of NHL repeat containing 2 (Nhlrc2) leads to early embryonic lethality in mice, but the exact timing is currently unknown. In this study, we determined the time of lethality for Nhlrc2 knockout (KO), C57BL/6NCrl-Nhlrc2tm1a(KOMP)Wtsi /Oulu, embryos and the in situ expression pattern of Nhlrc2 based on LacZ reporter gene expression during this period. Nhlrc2 KO preimplantation mouse embryos developed normally after in vitro fertilization. Embryonic stem (ES) cells established from KO blastocysts proliferated normally despite a complete loss of the NHLRC2 protein. Nhlrc2 KO embryos from timed matings implanted and were indistinguishable from their wildtype littermates on embryonic day (E) 6.5. On E7.5, Nhlrc2 KO embryo development was arrested, and on E8.5, only 6% of the genotyped embryos were homozygous for the Nhlrc2tm1a(KOMP)Wtsi allele. Nhlrc2 KO E8.5 embryos showed limited embryonic or extraembryonic tissue differentiation and remained at the cylinder stage. Nhlrc2 expression was ubiquitous but strongest in the epiblast/ectoderm and extraembryonic ectoderm on E6.5 and E7.5. NHLRC2 is essential for early postimplantation development, and its loss leads to failed gastrulation and amniotic folding in mice. Future studies on the evolutionarily conserved NHLRC2 will provide new insights into the molecular pathways involved in the early steps of postimplantation development.


Subject(s)
Gastrulation , Germ Layers , Animals , Cell Differentiation/genetics , Ectoderm , Gastrulation/genetics , Mice , Mice, Inbred C57BL
2.
Respir Res ; 23(1): 206, 2022 Aug 13.
Article in English | MEDLINE | ID: mdl-35964085

ABSTRACT

BACKGROUND: Variants of NHL repeat-containing protein 2 (NHLRC2) have been associated with severe fibrotic interstitial lung disease in early childhood and NHLRC2 has been listed as a differentially expressed gene between rapidly and slowly progressing idiopathic pulmonary fibrosis (IPF) patients. However, its cell type-specific localization in human lung tissue is unknown. The aim of this study was to evaluate NHLRC2 mRNA and protein expression in different cell types of lung tissue samples and to investigate the effect of transforming growth factor (TGF)-ß1 exposure on NHLRC2 expression in vitro. METHODS: The NHLRC2 expression in lung tissue samples was studied by immunohistochemistry (50 IPF, 10 controls) and mRNA in situ hybridization (8 IPF, 3 controls). The immunohistochemical NHLRC2 expression was quantified with image analysis software and associated with the clinical and smoking data of the patients. NHLRC2 expression levels in primary stromal and small airway epithelial cell lines after exposure to TGF-ß1 was measured by quantitative reverse transcription polymerase chain reaction and Western blot analysis. RESULTS: NHLRC2 expression was detected especially in bronchiolar epithelial cells, type II pneumocytes and macrophages in normal lung. In the lungs of IPF patients, NHLRC2 was mainly expressed in hyperplastic alveolar epithelial cells lining fibroblast foci and honeycombs. NHLRC2 expression assessed by image analysis was higher in IPF compared to controls (p < 0.001). Ever-smokers had more prominent NHLRC2 staining than non-smokers (p = 0.037) among IPF patients. TGF-ß1 exposure did not influence NHLRC2 levels in lung cell lines. CONCLUSIONS: NHLRC2 expression was higher in IPF compared to controls being widely expressed in type II pneumocytes, macrophages, bronchiolar epithelium, and hyperplastic alveolar epithelium. Additionally, its expression was not regulated by the exposure to TGF-ß1 in vitro. Further studies are needed to clarify the role of NHLRC2 in IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis , Intracellular Signaling Peptides and Proteins/metabolism , Child, Preschool , Fibroblasts/metabolism , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , RNA, Messenger/genetics , Transforming Growth Factor beta1/metabolism
3.
Ultrastruct Pathol ; 45(1): 37-48, 2021 Jan 02.
Article in English | MEDLINE | ID: mdl-33377815

ABSTRACT

This study aimed at an ultrastructural characterization of myofibroblasts cultured from different compartments of lung from never-smokers and smokers with or without COPD. In addition, we evaluated the expression of alpha smooth muscle actin (α-SMA), a marker for myofibroblasts, and contractile properties. Stromal cells cultured from central and corresponding peripheral or only from peripheral lung of never-smokers, smokers without COPD and COPD patients were analyzed by transmission electron microscopy (TEM), immunoelectron microscopy (IEM), Western analysis and/or by collagen gel contraction assay. TEM revealed that myofibroblasts cultured from smokers and COPD had less prominent intracellular actin filaments. We also examined fibronexus (FNX), which is a typical ultrastructural feature of myofibroblasts, and observed that patients with COPD more frequently had tandem-like FNX as compared to other samples. Western analysis showed that the samples derived from the central lung of never-smokers expressed higher levels of α-SMA than those of smokers and COPD patients. Cells from central lung were less contractile than those from peripheral lung. We conclude that myofibroblasts have variable ultrastructural and functional properties based on their localization in the lung and, moreover, these properties are affected by both smoking history and COPD.


Subject(s)
Fibroblasts , Myofibroblasts , Humans , Lung , Microscopy, Immunoelectron , Smoking/adverse effects
4.
Hum Mol Genet ; 27(24): 4288-4302, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30239752

ABSTRACT

The development of tissue fibrosis is complex and at the present time, not fully understood. Fibrosis, neurodegeneration and cerebral angiomatosis (FINCA disease) have been described in patients with mutations in NHL repeat-containing protein 2 (NHLRC2). However, the molecular functions of NHLRC2 are uncharacterized. Herein, we identified putative interacting partners for NHLRC2 using proximity-labeling mass spectrometry. We also investigated the function of NHLRC2 using immortalized cells cultured from skin biopsies of FINCA patients and normal fibroblasts with NHLRC2 knock-down and NHLRC2 overexpressing gene modifications. Transmission electron microscopy analysis of immortalized cell cultures from three FINCA patients demonstrated multilamellar bodies and distinctly organized vimentin filaments. Additionally, two of three cultures derived from patient skin biopsies contained cells that exhibited features characteristic of myofibroblasts. Altogether, the data presented in this study show for the first time that NHLRC2 is involved in cellular organization through regulation of the cytoskeleton and vesicle transport. We conclude that compound heterozygous p.Asp148Tyr and p.Arg201GlyfsTer6 mutations in NHLRC2 lead to severe tissue fibrosis in humans by enhancing the differentiation of fibroblasts to myofibroblasts.


Subject(s)
Angiomatosis/pathology , Brain Diseases/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Myofibroblasts/pathology , Nerve Degeneration/genetics , Actins/genetics , Angiomatosis/genetics , Brain Diseases/genetics , Cell Differentiation/genetics , Cells, Cultured , Fibrosis , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mutation/genetics , Myofibroblasts/metabolism , Skin/metabolism , Skin/pathology
5.
Biol Reprod ; 102(2): 306-315, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31494675

ABSTRACT

Stanniocalcin-1 (STC-1) is a pro-survival factor that protects tissues against stressors, such as hypoxia and inflammation. STC-1 is co-expressed with the endometrial receptivity markers, and recently endometrial STC-1 was reported to be dysregulated in endometriosis, a condition linked with endometrial progesterone resistance and inflammation. These features are also common in the endometrium in women with polycystic ovary syndrome (PCOS), the most common endocrine disorder in women. Given that women with PCOS present with subfertility, pregnancy complications, and increased risk for endometrial cancer, we investigated endometrial STC-1 expression in affected women. Endometrial biopsy samples were obtained from women with PCOS and controls, including samples from overweight/obese women with PCOS before and after a 3-month lifestyle intervention. A total of 98 PCOS and 85 control samples were used in immunohistochemistry, reverse-transcription polymerase chain reaction, or in vitro cell culture. STC-1 expression was analyzed at different cycle phases and in endometrial stromal cells (eSCs) after steroid hormone exposure. The eSCs were also challenged with 8-bromo-cAMP and hypoxia for STC-1 expression. The findings indicate that STC-1 expression is not steroid hormone mediated although secretory-phase STC-1 expression was blunted in PCOS. Lower expression seems to be related to attenuated STC-1 response to stressors in PCOS eSCs, shown as downregulation of protein kinase A activity. The 3-month lifestyle intervention did not restore STC-1 expression in PCOS endometrium. More studies are warranted to further elucidate the mechanisms behind the altered endometrial STC-1 expression and rescue mechanism in the PCOS endometrium.


Subject(s)
Endometrium/metabolism , Glycoproteins/metabolism , Overweight/metabolism , Polycystic Ovary Syndrome/metabolism , Adult , Cell Cycle/physiology , Female , Glycoproteins/genetics , Humans , Obesity/genetics , Obesity/metabolism , Overweight/genetics , Polycystic Ovary Syndrome/genetics , Stromal Cells/metabolism
6.
Tumour Biol ; 37(10): 13811-13820, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27481516

ABSTRACT

Mucin-1 (MUC1) affects cancer progression in lung adenocarcinoma, and its aberrant expression pattern has been correlated with poor tumor differentiation and impaired prognosis. In this study, the immunohistochemical expression of MUC1 and Mucin-4 (MUC4) was analyzed in a series of 106 surgically operated stage I-IV pulmonary adenocarcinomas. MUC1 immunohistochemistry was evaluated according to the Nagai classification, and the immunohistochemical profile of the tumors was correlated with detailed clinical and histological data. The effect of cigarette smoke on MUC1 expression in lung cancer cell lines was examined using real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) and immunoelectron microscopy (IEM). In contrast to the normal apical localization of MUC1, a basolateral and cytoplasmic (depolarized) MUC1 expression pattern was frequently encountered in the high-grade subtypes, i.e., solid predominant adenocarcinoma and the cribriform variant of acinar predominant adenocarcinoma (p < 0.001), and was rarely observed in tumors containing a non-predominant lepidic component (p < 0.001). Furthermore, the altered staining pattern of MUC1 correlated with stage (p = 0.002), reduced overall survival (p = 0.031), and was associated with smoking (p < 0.001). When H1650 adenocarcinoma cells were exposed to cigarette smoke and analyzed by RT-qPCR and IEM, the levels of the MUC1 transcript and protein were elevated (p = 0.042). In conclusion, MUC1 participates in the pathogenesis of lung adenocarcinoma and associates with smoking both in vitro and in vivo. In lung adenocarcinoma, depolarized MUC1 protein expression correlated with histological growth patterns, stage, and patient outcome.


Subject(s)
Adenocarcinoma/mortality , Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Mucin-1/metabolism , Smoking/adverse effects , Adenocarcinoma/etiology , Adenocarcinoma/metabolism , Aged , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lung Neoplasms/etiology , Lung Neoplasms/metabolism , Male , Microscopy, Immunoelectron , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured
7.
Respir Res ; 17: 14, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26846335

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is an incurable lung disease with a poor prognosis. Fibroblasts and myofibroblasts are the key cells in the fibrotic process. Recently two drugs, pirfenidone and nintedanib, were approved for clinical use as they are able to slow down the disease progression. The mechanisms by which these two drugs act in in vitro cell systems are not known. The aim of this study was therefore to examine the effects of pirfenidone and nintedanib on fibroblasts and myofibroblasts structure and function established from patients with or without IPF. METHODS: Stromal cells were collected and cultured from control lung (n = 4) or IPF (n = 7). The cells were treated with pirfenidone and/or nintedanib and the effect of treatment was evaluated by measuring cell proliferation, alpha smooth muscle actin (α-SMA) and fibronectin expression by Western analysis and/or immunoelectron microscopy, ultrastructural properties by transmission electron microscopy and functional properties by collagen gel contraction and invasion assays. RESULTS: Both pirfenidone and nintedanib reduced in vitro proliferation of fibroblastic cells in a dose dependent manner. The number of cells from control lung was reduced to 47 % (p = 0.04) and of IPF cells to 42 % (p = 0.04) by 1 mM pirfenidone and correspondingly to 67 % (p = 0.04) and 68 % (p = 0.04), by 1 µM nintedanib. If both drugs were used together, a further reduced proliferation was observed. Both pirfenidone and nintedanib were able to reduce the amount of α-SMA and the myofibroblastic appearance although the level of reduction was cell line dependent. In functional assays, the effect of both drugs was also variable. CONCLUSIONS: We conclude that the ultrastructure and function of fibroblasts and myofibroblasts are affected by pirfenidone and nintedanib. Combination of the drugs reduced cell proliferation more than either of them individually. Human lung derived cell culture systems represent a potential platform for screening and testing drugs for fibrotic diseases.


Subject(s)
Fibroblasts/drug effects , Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Indoles/administration & dosage , Pyridones/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Therapy, Combination , Humans , Myofibroblasts/drug effects , Myofibroblasts/pathology , Treatment Outcome
8.
Oncology ; 88(4): 214-25, 2015.
Article in English | MEDLINE | ID: mdl-25502907

ABSTRACT

The interplay between tumor stroma and breast cancer cells (BCCs) is thought to play a significant role in breast cancer. The current knowledge of human mesenchymal stromal cell (MSC) and BCC interaction is contradictory, and the donor sex issue is not addressed at all. We hypothesized that donor sex could have an effect on proliferation of MSCs or BCCs in co-culture in vitro. Three estrogen receptor-negative BCC lines, 19 primary human MSCs and breast tissue-derived fibroblasts from 4 donors were used. MSCs from female donors enhanced BCC proliferation (p = 0.005). The change in BCC proliferation was only partly due to soluble factors excreted by MSCs. The highly aggressive BCC line MDA-MB- 231 induced the proliferation of MSCs (p < 0.001) and fibroblasts (p = 0.037) in co-culture experiments. The magnitude in proliferation change was cell line dependent and partly sex dependent.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation , Mesenchymal Stem Cells/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Breast/pathology , Cell Differentiation , Cell Movement , Coculture Techniques , Female , Fibroblasts/pathology , Flow Cytometry , Humans , Male , Middle Aged , Neoplasm Invasiveness , Sex Factors , Tumor Cells, Cultured , Young Adult
9.
Scand Cardiovasc J ; 49(2): 82-94, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25705991

ABSTRACT

OBJECTIVE: The clinical data considering the bone marrow mononuclear cell (BMMNC) therapy in treatment for acute myocardial infarction (AMI) are controversial and the mechanisms remain unknown. Our objective was to study the cardiac function and changes in cytokine levels after administration of BMMNC in experimental AMI model. DESIGN: Unlabeled or Super-Paramagnetic-Iron-Oxide-labeled BMMNCs or saline was injected into myocardium of 31 pigs after circumflex artery occlusion. Ejection fraction (EF) was measured preoperatively, postoperatively and at 21 days by echocardiography. Cardiac MRI was performed postoperatively and after 21 days in 7 BMMNC animals. Serum cytokine levels were measured at baseline, 24 h and 21 days. Cellular homing was evaluated comparing MRI and histology. RESULTS: From baseline to 21 days EF decreased less in BMMNC group (EF mean control -19 SD 12 vs. BMMNC -4 SD 15 percentage points p = 0.02). Cytokine concentrations showed high variability between the animals. MRI correlated with histology in cell detection and revealed BMMNCs in the infarction area. By MRI, EF improved 11 percentage points. The improvement in EF was associated with the number of transplanted BMMNCs detected in the myocardium. CONCLUSION: BMMNC injection after AMI improved cardiac function. Quantity of transplanted BMMNCs correlated with the improvement in cardiac function after AMI.


Subject(s)
Bone Marrow Transplantation , Myocardial Infarction/therapy , Stem Cell Transplantation , Animals , Disease Models, Animal , Magnetic Resonance Imaging , Myocardium/pathology , Stroke Volume , Swine
10.
Stem Cells ; 31(2): 317-26, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23132820

ABSTRACT

The promising clinical effects of mesenchymal stromal/stem cells (MSCs) rely especially on paracrine and nonimmunogenic mechanisms. Delivery routes are essential for the efficacy of cell therapy and systemic delivery by infusion is the obvious goal for many forms of MSC therapy. Lung adhesion of MSCs might, however, be a major obstacle yet to overcome. Current knowledge does not allow us to make sound conclusions whether MSC lung entrapment is harmful or beneficial, and thus we wanted to explore MSC lung adhesion in greater detail. We found a striking difference in the lung clearance rate of systemically infused MSCs derived from two different clinical sources, namely bone marrow (BM-MSCs) and umbilical cord blood (UCB-MSCs). The BM-MSCs and UCB-MSCs used in this study differed in cell size, but our results also indicated other mechanisms behind the lung adherence. A detailed analysis of the cell surface profiles revealed differences in the expression of relevant adhesion molecules. The UCB-MSCs had higher expression levels of α4 integrin (CD49d, VLA-4), α6 integrin (CD49f, VLA-6), and the hepatocyte growth factor receptor (c-Met) and a higher general fucosylation level. Strikingly, the level of CD49d and CD49f expression could be functionally linked with the lung clearance rate. Additionally, we saw a possible link between MSC lung adherence and higher fibronectin expression and we show that the expression of fibronectin increases with MSC culture confluence. Future studies should aim at developing methods of transiently modifying the cell surface structures in order to improve the delivery of therapeutic cells.


Subject(s)
Bone Marrow Cells/cytology , Cord Blood Stem Cell Transplantation , Fetal Blood/cytology , Lung/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Biomarkers/metabolism , Bone Marrow Cells/metabolism , Cell Adhesion , Cell Differentiation , Female , Fetal Blood/metabolism , Gene Expression , Half-Life , Humans , Infusions, Intravenous , Integrin alpha4/genetics , Integrin alpha4/metabolism , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Integrin alpha6/genetics , Integrin alpha6/metabolism , Integrin alpha6beta1/genetics , Integrin alpha6beta1/metabolism , Isotope Labeling , Lung/immunology , Lung/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Technetium Compounds , Transplantation, Heterologous
11.
Respir Res ; 14: 84, 2013 Aug 11.
Article in English | MEDLINE | ID: mdl-23937155

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is characterized by structural changes in alveoli and airways. Our aim was to analyse the numbers of alpha-smooth muscle actin (α-SMA) positive cells, as a marker of myofibroblasts, in different lung compartments in non-smokers and smokers with normal lung function or COPD. METHODS: α-SMA, tenascin-C (Tn-C) and EDA-fibronectin in alveolar level and airways were assayed by immunohistochemistry and quantified by image analysis. Immunohistochemical findings were correlated with clinical data. α-SMA protein was also analysed by Western blotting from fibroblastic cells cultured from peripheral lung of non-smokers, smokers without COPD and smokers with COPD. RESULTS: In many cases, the endings of the detached alveolar walls were widened, the structures of which were named as widened alveolar tips. Widened alveolar tips contained α-SMA positive cells, which were obviously myofibroblasts. There were less alveolar tips containing positive cells for α-SMA in alveoli and α-SMA positive cells in bronchioles in smokers and in COPD compared to non-smokers. The quantity of α-SMA positive cells was increased in bronchi in COPD. Tn-C was elevated in bronchi in COPD and smokers' lung. The α-SMA protein level was 1.43-fold higher in stromal cells cultured from non-smokers than in those of smokers. CONCLUSIONS: Myofibroblasts are localized variably in normal and diseased lung. This indicates that they have roles in both regeneration of lung and pathogenesis of COPD. The widened alveolar tips, these newly characterized histological structures, seemed to be the source of myofibroblasts at the alveolar level.


Subject(s)
Lung/pathology , Myofibroblasts/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Smoking/pathology , Aged , Cells, Cultured , Cytokines/immunology , Female , Humans , Lung/immunology , Male , Middle Aged , Myofibroblasts/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Smoking/immunology
12.
FASEB J ; 26(11): 4400-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22798425

ABSTRACT

Adipose tissue inflammation is an important pathological process in obese people, associated with diabetes and cardiovascular disease. We hypothesized that 1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] inhibits cytokine secretion from adipocytes via direct inhibition of transcription factor nuclear factor-κB (NF-κB). We utilized two different human models. Bone marrow-derived human mesenchymal stromal cells (hMSCs) differentiated into adipocytes, and adipocytes isolated from biopsies stimulated with lipopolysaccharide (LPS) were treated with or without 1,25(OH)(2)D(3). Expression and secretion of interleukin-6 (IL-6) were measured by quantitative RT-PCR analysis and ELISA. Assessment of NF-κB nuclear translocation, DNA binding activity was performed by immunofluorescence (IF) and electrophoretic mobility assay (EMSA). Inhibitor κB (IκB) and its phosphorylation were detected by Western blot (WB) analysis. Simultaneous 1,25(OH)(2)D(3) cotreatment significantly reduced LPS-stimulated (10 ng/ml) IL-6 secretion dose dependently by 15% at 10(-10) M and 26% at 10(-7) M (P<0.05) in hMSCs, while preincubation with 1,25(OH)(2)D(3) (10(-7) M) for 24 h reduced IL-6 secretion by 24 and 35% (P<0.001) and mRNA levels by 34 and 30% (P<0.05) in hMSCs and isolated adipocytes, respectively. 1,25(OH)(2)D(3) suppressed LPS-stimulated IκB phosphorylation-mediated NF-κB translocation into the nucleus were evident from WB, IF, and EMSA. 1,25(OH)(2)D(3) inhibits LPS-stimulated IL-6 secretion in two human adipocyte models via interference with NF-κB signaling.


Subject(s)
Adipocytes/metabolism , Calcitriol/pharmacology , Cytokines/metabolism , Gene Expression Regulation/drug effects , NF-kappa B/metabolism , Adipocytes/drug effects , Adolescent , Bone Marrow Cells/cytology , Cells, Cultured , Cytokines/genetics , Female , Humans , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Mesenchymal Stem Cells/cytology , Middle Aged , NF-kappa B/genetics
13.
Scand Cardiovasc J ; 47(2): 114-20, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23157430

ABSTRACT

OBJECTIVES: Bone marrow mononuclear cells (BM-MNCs) can ameliorate focal ischaemic brain injury. A global ischaemic brain injury, which can occur after cardiac or thoracic surgery, could be an essential target for BM-MNCs. No studies using BM-MNCs for this indication have been conducted. DESIGN: Ten porcine underwent a global normothermic ischaemic insult, followed by an intra-arterial injection of Technetium(99m)-HMPAO-labelled BM-MNCs after 2, 4, 6, 12 or 24 hours. A whole-body scan and a SPECT/CT were performed 2 hours after the injection. Severity of the injury was assessed with EEG and tissue biopsies were analysed by scintigraphy. RESULTS: The majority of the cells appeared in the lungs and the liver. Only a minimal number of cells were located in the brain. Median distribution of cells between organs in all animals was as follows: lungs 32.7% (30.6-38.2), liver 14.2% (12.0-17.2), spleen 7.3% (3.3-11.3) and kidneys 2.5% (2.0-3.3). The transplanted cells could not be detected within the brain tissue by radionuclide imaging. CONCLUSIONS: Intra-arterially transplanted BM-MNCs did not migrate to the damaged brain tissue in significant quantity when transplanted during the first 24 hours after the global ischaemic insult, contrary to results with models of focal brain injury.


Subject(s)
Bone Marrow Cells , Bone Marrow Transplantation , Brain Ischemia/diagnostic imaging , Brain Ischemia/therapy , Injections, Intra-Arterial , Animals , Biopsy , Brain Ischemia/pathology , Disease Models, Animal , Electroencephalography , Injury Severity Score , Monocytes/cytology , Multimodal Imaging , Positron-Emission Tomography , Sus scrofa , Technetium , Tissue Distribution , Tomography, X-Ray Computed
14.
Transl Lung Cancer Res ; 12(6): 1221-1235, 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37425408

ABSTRACT

Background: Certain variants of NHL repeat (named after NCL-1, HT2A and LIN-41)-containing protein 2 (NHLRC2) gene have been linked to severe fibrotic interstitial lung disease in children. The aim of the current study was to evaluate the expression of NHLRC2 in lung cell and tissue samples from patients with lung adenocarcinoma (ADC) and squamous cell carcinoma (SCC). Methods: The expression of NHLRC2 in lung tissue samples was studied by immunohistochemistry (102 ADC, 111 SCC), mRNA in situ hybridization (4 ADC, 3 SCC), and Western blot analysis (3 ADC, 2 SCC). The immunohistochemical NHLRC2 expression was measured by image analysis software and the percentage of NHLRC2-positive cancer cells was evaluated by semiquantitative analysis. The immunohistochemical results of NHLRC2 were compared with the clinical and histological characteristics of the patients. NHLRC2 protein levels in primary stromal and epithelial lung cancer cell lines were measured by Western blot analysis. Results: NHLRC2 was mainly expressed in cancer cells and inflammatory cells within the tumor. The NHLRC2 expression evaluated by image analysis method was significantly higher in ADC compared with that in SCC (P<0.001). High NHLRC2 expression was associated with reduced disease specific survival (P=0.002), overall survival (P=0.001), and high mitotic activity (P=0.042) in ADC. Additionally, the proportion of NHLRC2-positive cancer cells analyzed by the semiquantitative method was significantly higher in ADC than in SCC (P<0.001). Conclusions: NHLRC2 expression was higher in lung ADC than in SCC and its expression was associated with poor survival in ADC patients. Further studies are required to clarify the pathogenetic role of NHLRC2 in lung cancer.

15.
Lab Invest ; 92(9): 1270-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22710982

ABSTRACT

The characteristic features of myofibroblasts in various lung disorders are poorly understood. We have evaluated the ultrastructure and invasive capacities of myofibroblasts cultured from small volumes of diagnostic bronchoalveolar lavage (BAL) fluid samples from patients with different types of lung diseases. Cells were cultured from samples of BAL fluid collected from 51 patients that had undergone bronchoscopy and BAL for diagnostic purposes. The cells were visualized by transmission electron microscopy and immunoelectron microscopy to achieve ultrastructural localization of alpha-smooth muscle actin (α-SMA) and fibronectin. The levels of α-SMA protein and mRNA and fibronectin mRNA were measured by western blot and quantitative real-time reverse transcriptase polymerase chain reaction. The invasive capacities of the cells were evaluated. The cultured cells were either fibroblasts or myofibroblasts. The structure of the fibronexus, and the amounts of intracellular actin, extracellular fibronectin and cell junctions of myofibroblasts varied in different diseases. In electron and immunoelectron microscopy, cells cultured from interstitial lung diseases (ILDs) expressed more actin filaments and α-SMA than normal lung. The invasive capacity of the cells obtained from patients with idiopathic pulmonary fibrosis was higher than that from patients with other type of ILDs. Cells expressing more actin filaments had a higher invasion capacity. It is concluded that electron and immunoelectron microscopic studies of myofibroblasts can reveal differential features in various diseases. An analysis of myofibroblasts cultured from diagnostic BAL fluid samples may represent a new kind of tool for diagnostics and research into lung diseases.


Subject(s)
Lung Diseases, Interstitial/pathology , Myofibroblasts/ultrastructure , Actins/metabolism , Base Sequence , Biopsy , Blotting, Western , Bronchoalveolar Lavage Fluid , DNA Primers , Humans , Microscopy, Electron, Transmission , Microscopy, Immunoelectron , Myofibroblasts/pathology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
Scand J Gastroenterol ; 47(11): 1375-85, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22954013

ABSTRACT

OBJECTIVE: Cytokines initiate and modify systemic inflammatory response in early acute pancreatitis. The aim of this study was to analyze which cytokines are released from the pancreas to portal venous blood in the early phase of acute experimental necrotizing and oedematous pancreatitis and which of those cytokines are correlated with the more severe form of the disease. MATERIAL AND METHODS: Fifteen pigs were randomized to develop mild oedematous pancreatitis (n = 5, saline infusion to pancreatic duct), severe necrotizing pancreatitis (n = 5, taurocholic acid infusion) along with a control group (n = 5). Arterial and venous blood samples were drawn and cytokine levels were measured from portal vein blood at 0, 120, 240 and 360 min after the induction of pancreatitis. Tissue samples from the pancreas were harvested at 0 and 360 min. RESULTS: White blood cell count increased in necrotizing pancreatitis and the control group. The amount of neutrophils increased (p < 0.001) and the lymphocyte and eosinophil counts decreased in all groups (p < 0.001, p < 0.001). The monocyte count, as well as PDGF and IL-6 concentrations, increased only in necrotizing pancreatitis. IL-8 and eotaxin increased both in oedematous and necrotizing pancreatitis. MCP-1 increased in all groups. IL-9, IL-4, MIP-1α, IFN- γ concentrations did not change. Eotaxin and MCP-1 plasma levels from a previous series between portal venous and pulmonary arterial blood were not significantly different. CONCLUSIONS: The initial inflammatory process was diverse in oedematous and necrotizing pancreatitis. Increased monocyte count in combination with elevated PDGF and IL-6 are characteristic of necrotizing pancreatitis in our model.


Subject(s)
Cytokines/blood , Edema/blood , Leukocyte Count , Pancreatitis, Acute Necrotizing/blood , Analysis of Variance , Animals , Disease Models, Animal , Edema/chemically induced , Edema/complications , Eosinophils , Lymphocyte Count , Monocytes , Neutrophils , Pancreatitis, Acute Necrotizing/chemically induced , Pancreatitis, Acute Necrotizing/complications , Platelet-Derived Growth Factor/metabolism , Portal Vein , Pulmonary Artery , Sodium Chloride , Swine , Taurocholic Acid , Time Factors
17.
Cells ; 11(14)2022 07 18.
Article in English | MEDLINE | ID: mdl-35883669

ABSTRACT

Bronchoalveolar lavage (BAL) is an important diagnostic and research tool for the investigation of various lung diseases. In addition to inflammatory and epithelial cells, BAL fluid may contain a small number of stromal cells, such as fibroblasts. During the past 30 years, a number of research groups have cultured BAL-derived fibroblasts for several passages in vitro. In addition to fibroblasts, these cultures have been reported to contain fibrocytes, myofibroblasts, and stem cells. We aim to present a summary of studies that have cultured stromal cells from BAL fluid.


Subject(s)
Lung Diseases, Interstitial , Therapeutic Irrigation , Bronchoalveolar Lavage , Cell Line , Fibroblasts/metabolism , Humans , Lung Diseases, Interstitial/metabolism
18.
Inflammation ; 45(2): 768-779, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34686945

ABSTRACT

Mast cells (MCs) are known to be involved in the pathogenesis of idiopathic pulmonary fibrosis (IPF), although their role in acute exacerbations of IPF has not been investigated. The aims of the study were to evaluate the numbers of MCs in fibrotic and non-fibrotic areas of lung tissue specimens of idiopathic pulmonary fibrosis (IPF) patients with or without an acute exacerbation of IPF, and to correlate the MC density with clinical parameters. MCs of IPF patients were quantified from surgical lung biopsy (SLB) specimens (n = 47) and lung tissue specimens taken at autopsy (n = 7). MC density was higher in the fibrotic areas of lung tissue compared with spared alveolar areas or in controls. Female gender, low diffusion capacity for carbon monoxide, diffuse alveolar damage, and smoking were associated with a low MC density. MC densities of fibrotic areas had declined significantly in five subjects in whom both SLB in the stable phase and autopsy after an acute exacerbation of IPF had been performed. There were no correlations of MC densities with survival time or future acute exacerbations. The MC density in fibrotic areas was associated with several clinical parameters. An acute exacerbation of IPF was associated with a significant decline in MC counts. Further investigations will be needed to clarify the role of these cells in IPF and in the pathogenesis of acute exacerbation as this may help to identify some potential targets for medical treatment for this serious disease.


Subject(s)
Idiopathic Pulmonary Fibrosis , Cell Count , Female , Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Mast Cells/pathology
19.
PLoS One ; 16(4): e0250109, 2021.
Article in English | MEDLINE | ID: mdl-33905434

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) and lung cancer share common risk factors, epigenetic and genetic alterations, the activation of similar signaling pathways and poor survival. The aim of this study was to examine the gene expression profiles of stromal cells from patients with IPF and lung adenocarcinoma (ADC) as well as from normal lung. The gene expression levels of cultured stromal cells derived from non-smoking patients with ADC from the tumor (n = 4) and the corresponding normal lung (n = 4) as well as from patients with IPF (n = 4) were investigated with Affymetrix microarrays. The expression of collagen type IV alpha 1 chain, periostin as well as matrix metalloproteinase-1 and -3 in stromal cells and lung tissues were examined with quantitative real-time reverse transcriptase polymerase chain reaction and immunohistochemistry, respectively. Twenty genes were similarly up- or down-regulated in IPF and ADC compared to control, while most of the altered genes in IPF and ADC were differently expressed, including several extracellular matrix genes. Collagen type IV alpha 1 chain as well as matrix metalloproteinases-1 and -3 were differentially expressed in IPF compared to ADC. Periostin was up-regulated in both IPF and ADC in comparison to control. All studied factors were localized by immunohistochemistry in stromal cells within fibroblast foci in IPF and stroma of ADC. Despite the similarities found in gene expressions of IPF and ADC, several differences were also detected, suggesting that the molecular changes occurring in these two lung illnesses are somewhat different.


Subject(s)
Adenocarcinoma of Lung/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Stromal Cells/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Aged , Cell Adhesion Molecules/genetics , Cells, Cultured , Collagen Type IV/genetics , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Female , Fibroblasts/metabolism , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 3/genetics , Middle Aged , Signal Transduction , Transcriptome/genetics
20.
BMC Dev Biol ; 10: 113, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21080917

ABSTRACT

BACKGROUND: The aim of this study was to analyze the cell-specific expression of E- and N-cadherin and ß-catenin in developing human lung tissues from 12 to 40 weeks of gestation. METHODS: Forty-seven cases of developing human lung including pseudoglandular, canalicular, saccular and alveolar periods were analyzed by immunohistochemistry for E- and N-cadherin and ß-catenin and twenty-one cases were also investigated by RT-PCR for E- and N-cadherin and ß-catenin. For identifying the lung cells, the sections were also stained with antibodies against thyroid transcription factor-1 (TTF-1) and caveolin-1. Normal adult lung tissue was used as a control. E-cadherin was strongly expressed in epithelium of bronchi and large bronchioles from week 12 onwards and it was also positive in alveoli in pretype II cells and type II cells. N-cadherin was present in most of the epithelial cells of bronchi and the largest bronchioles during the pseudo-glandular and canalicular periods. N-cadherin was not detected in epithelium of developing alveoli. ß-catenin was strongly membrane-bound and positively expressed in bronchial epithelium from week 12 to week 40; it showed nuclear positivity in both developing airway epithelium and in the cells underneath the epithelium during pseudo-glandular period and to a lesser degree also in the canalicular period. ß-catenin was positive in pretype II cells as well as in type I and type II pneumocytes within alveoli. RT-PCR analyses revealed detectable amounts of RNAs of E- and N-cadherin and ß-catenin in all cases studied. The amounts of RNAs were higher in early stages of gestation. CONCLUSIONS: E-cadherin is widely expressed in bronchial and alveolar epithelial cells. N-cadherin exhibit extensive epithelial positivity in bronchial epithelial cells during early lung development. The presence of ß-catenin was observed in several cell types with a distinct location in tissue and cells in various gestational stages, indicating that it possesses several roles during lung development. The expressions of protein and mRNAs of E- and N-cadherin and ß-catenin were higher in early gestation compared to of the end. Moreover, the expressions of these factors were higher during the lung development than in the adult human lung.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Epithelial Cells/physiology , Lung/embryology , Up-Regulation , beta Catenin/metabolism , Adult , Antigens, CD/genetics , Cadherins/genetics , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Female , Fetus/anatomy & histology , Fetus/physiology , Gestational Age , Humans , Infant , Lung/anatomy & histology , Lung/metabolism , Pregnancy , Respiratory Mucosa/cytology , Retrospective Studies , Transcription Factors , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL