Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Invest Ophthalmol Vis Sci ; 63(1): 27, 2022 01 03.
Article in English | MEDLINE | ID: mdl-35060996

ABSTRACT

Purpose: Erianin has been reported to inhibit tumor activity by suppressing the expression of integrins. It is hypothesized that erianin can inhibit retinal neovascularization in collagen by suppressing the expression of integrins. With an aim to test this hypothesis, the regulation of erianin on collagen-mediated retinal angiogenesis via the Ras homolog gene family member A (RhoA)/Rho-associated coiled-coil containing protein kinase 1 (ROCK1) signaling pathway induced by α2 and ß1 integrin-collagen interactions was investigated. Methods: The effects of erianin on human retinal vascular endothelial cells (HRVECs) were assessed in vitro using a hypoxia model in a three-dimensional cell culture induced by cobalt (II) chloride (CoCl2). A hypoxia-induced retinopathy model in adult zebrafish and zebrafish embryos was established to assess the antiangiogenic effect of erianin with and without vitreous collagen in vivo. The expression of α2 and ß1 integrin and RhoA/ROCK1 pathway in HRVECs and zebrafish retinas were analyzed. Results: In vitro, collagen improved the angiogenic potential of HRVECs, including migration, adhesion, and tube formation, in a three-dimensional cell culture model. Erianin suppressed the angiogenic processes of the CoCl2-induced hypoxia HRVEC model in a concentration-dependent manner. In vivo, erianin reduced retinal angiogenesis in the hypoxia-induced retinopathy model in adult and embryo zebrafish. Erianin inhibited the expression of α2 and ß1 integrin and RhoA/ROCK1 in a hypoxia-induced model in vitro in three-dimensional cell culture and in vivo in adult zebrafish. Conclusions: Collagen-mediated retinal angiogenesis may be regulated by erianin via the RhoA/ROCK1 signaling pathway induced by α2 and ß1 integrin-collagen interactions. These findings suggest that erianin has the therapeutic potential on intraocular collagen-mediated retinal angiogenesis.


Subject(s)
Bibenzyls/pharmacology , Gene Expression Regulation , Integrin alpha1/genetics , Integrin beta1/genetics , Monomeric GTP-Binding Proteins/genetics , Phenol/pharmacology , Retinal Neovascularization/genetics , Zebrafish Proteins/genetics , rho-Associated Kinases/genetics , Animals , Cell Movement , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Integrin alpha1/biosynthesis , Integrin beta1/biosynthesis , Monomeric GTP-Binding Proteins/biosynthesis , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Signal Transduction , Zebrafish , Zebrafish Proteins/biosynthesis , rho-Associated Kinases/biosynthesis
2.
Front Endocrinol (Lausanne) ; 12: 635405, 2021.
Article in English | MEDLINE | ID: mdl-34025576

ABSTRACT

Background: Cell therapy of diabetes aims at restoring the physiological control of blood glucose by transplantation of functional pancreatic islet cells. A potentially unlimited source of cells for such transplantations would be islet cells derived from an in vitro differentiation of human pluripotent stem cells (hESC/hiPSC). The islet-like clusters (ILC) produced by the known differentiation protocols contain various cell populations. Among these, the ß-cells that express both insulin and the transcription factor Nkx6.1 seem to be the most efficient to restore normoglycemia in diabetes animal models. Our aim was to find markers allowing selection of these efficient cells. Methods: Functional Cell-Capture Screening (FCCS) was used to identify markers that preferentially capture the cells expressing both insulin and Nkx6.1, from hESC-derived ILC cells. In order to test whether selection for such markers could improve cell therapy in diabetic mouse models, we used ILC produced from a clinical-grade line of hESC by a refined differentiation protocol adapted to up-scalable bioreactors. Re-aggregated MACS sorted cells were encapsulated in microspheres made of alginate modified to reduce foreign body reaction. Implantation was done intraperitoneally in STZ-treated C57BL/6 immuno-competent mice. Results: CD49A (integrin alpha1) was identified by FCCS as a marker for cells that express insulin (or C-peptide) as well as Nkx6.1 in ILC derived by hESC differentiation. The ILC fraction enriched in CD49A + cells rapidly reduced glycemia when implanted in diabetic mice, whereas mice receiving the CD49A depleted population remained highly diabetic. CD49A-enriched ILC cells also produced higher levels of human C-peptide in the blood of transplanted mice. However, the difference between CD49A-enriched and total ILC cells remained small. Another marker, CD26 (DPP4), was identified by FCCS as binding insulin-expressing cells which are Nkx6.1 negative. Depletion of CD26 + cells followed by enrichment for CD49A + cells increased insulin+/Nkx6.1+ cells fraction to ~70%. The CD26 - /CD49A + enriched ILC exhibited improved function over non-sorted ILC or CD49A + cells in diabetic mice and maintain prolonged blood C-peptide levels. Conclusions: Refining the composition of ILC differentiated from hPSC by negative selection to remove cells expressing CD26 and positive selection for CD49A expressing cells could enable more effective cell therapy of diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Dipeptidyl Peptidase 4/biosynthesis , Integrin alpha1/biosynthesis , Islets of Langerhans Transplantation , Islets of Langerhans/metabolism , Pluripotent Stem Cells/metabolism , Animals , C-Peptide/biosynthesis , Cell Differentiation , Cell Separation , Homeodomain Proteins/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Mice , Mice, Inbred C57BL , Microspheres
3.
Immunity ; 46(2): 287-300, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28214226

ABSTRACT

Tissue-resident memory T (Trm) cells form a heterogeneous population that provides localized protection against pathogens. Here, we identify CD49a as a marker that differentiates CD8+ Trm cells on a compartmental and functional basis. In human skin epithelia, CD8+CD49a+ Trm cells produced interferon-γ, whereas CD8+CD49a- Trm cells produced interleukin-17 (IL-17). In addition, CD8+CD49a+ Trm cells from healthy skin rapidly induced the expression of the effector molecules perforin and granzyme B when stimulated with IL-15, thereby promoting a strong cytotoxic response. In skin from patients with vitiligo, where melanocytes are eradicated locally, CD8+CD49a+ Trm cells that constitutively expressed perforin and granzyme B accumulated both in the epidermis and dermis. Conversely, CD8+CD49a- Trm cells from psoriasis lesions predominantly generated IL-17 responses that promote local inflammation in this skin disease. Overall, CD49a expression delineates CD8+ Trm cell specialization in human epithelial barriers and correlates with the effector cell balance found in distinct inflammatory skin diseases.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Integrin alpha1/immunology , Skin/immunology , T-Lymphocyte Subsets/immunology , Cell Separation , Flow Cytometry , Humans , Immunologic Memory/immunology , Integrin alpha1/biosynthesis , Lymphocyte Activation/immunology , Microscopy, Confocal , Psoriasis/immunology , Vitiligo/immunology
4.
Oncotarget ; 7(9): 10228-42, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26849234

ABSTRACT

Neuromedin U (NMU) was originally named based on its strong uterine contractile activity, but little is known regarding its signaling/functions in utero. We identified that NMU and one of its receptors, NMUR2, are not only present in normal uterine endometrium but also co-expressed in endometrial cancer tissues, where the NMU level is correlated with the malignant grades and survival of patients. Cell-based assays further confirmed that NMU signaling can promote cell motility and proliferation of endometrial cancer cells derived from grade II tumors. Activation of NMU pathway in these endometrial cancer cells is required in order to sustain expression of various adhesion molecules, such as CD44 and integrin alpha1, as well as production of their corresponding extracellular matrix ligands, hyaluronan and collagen IV; it also increased the activity of SRC and its downstream proteins RHOA and RAC1. Thus, it is concluded that NMU pathway positively controls the adhesion signaling-SRC-Rho GTPase axis in the tested endometrial cancer cells and that changes in cell motility and proliferation can occur when there is manipulation of NMU signaling in these cells either in vitro or in vivo. Intriguingly, this novel mechanism also explains how NMU signaling promotes the EGFR-driven and TGFß receptor-driven mesenchymal transitions. Through the above axis, NMU signaling not only can promote malignancy of the tested endometrial cancer cells directly, but also helps these cells to become more sensitive to niche growth factors in their microenvironment.


Subject(s)
Cell Transformation, Neoplastic/pathology , Endometrial Neoplasms/pathology , Endometrium/pathology , Neuropeptides/metabolism , Signal Transduction/physiology , Animals , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Collagen/biosynthesis , Disease Progression , Endometrium/metabolism , Female , Humans , Hyaluronan Receptors/biosynthesis , Hyaluronic Acid/biosynthesis , Integrin alpha1/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Nude , Middle Aged , Receptors, Neurotransmitter/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
5.
Oncotarget ; 7(18): 25194-207, 2016 May 03.
Article in English | MEDLINE | ID: mdl-26910370

ABSTRACT

A cell line was established from ventral prostate (VP) tumors of one-year-old Hi-Myc mice. These cells, called HMVP2 cells, are LinnegSca-1highCD49fhigh with high CD44 and CD29 expression and express CK14, Sca-1 and CD49f (but not CK8), suggesting basal-epithelial characteristics. Furthermore, HMVP2 cells form spheroids and both the cells and spheroids produce tumors in syngeneic mice. After four days of culture, HMVP2 spheroids underwent a gradual transition from LinnegSca-1highCD49fhigh expression to LinnegSca-1lowCD49flow while a subpopulation of the cells retained the original LinnegSca-1highCD49fhigh expression pattern. Additional cell subpopulations expressing Lin positive markers were also present suggesting further differentiation of HMVP2 spheroids. Two additional highly tumorigenic cell lines (HMVP2A1 and HMVP2A2) were isolated from HMVP2 cells after subsequent tumor formation in FVB/N mice. Concurrently, we also established cell lines from the VP of 6 months old Hi-Myc mice (named as HMVP1) and FVB/N mice (called NMVP) having less aggressive growth properties compared to the other three cell lines. AR expression was reduced in HMVP2 cells compared to NMVP and HMVP1 cells and almost absent in HMVP2A1 and HMVP2A2 cells. These cell lines will provide valuable tools for further mechanistic studies as well as preclinical studies to evaluate preventive and/or therapeutic agents for prostate cancer.


Subject(s)
Cell Line, Tumor , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology , Animals , Antigens, Ly/biosynthesis , Biomarkers, Tumor/metabolism , Cell Differentiation/physiology , Disease Models, Animal , Integrin alpha1/biosynthesis , Male , Membrane Proteins/biosynthesis , Mice , Mice, Transgenic
6.
J Biol Chem ; 290(10): 6546-57, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25593319

ABSTRACT

Hepatic insulin resistance is associated with increased collagen. Integrin α1ß1 is a collagen-binding receptor expressed on hepatocytes. Here, we show that expression of the α1 subunit is increased in hepatocytes isolated from high fat (HF)-fed mice. To determine whether the integrin α1 subunit protects against impairments in hepatic glucose metabolism, we analyzed glucose tolerance and insulin sensitivity in HF-fed integrin α1-null (itga1(-/-)) and wild-type (itga1(+/+)) littermates. Using the insulin clamp, we found that insulin-stimulated hepatic glucose production was suppressed by ∼50% in HF-fed itga1(+/+) mice. In contrast, it was not suppressed in HF-fed itga1(-/-) mice, indicating severe hepatic insulin resistance. This was associated with decreased hepatic insulin signaling in HF-fed itga1(-/-) mice. Interestingly, hepatic triglyceride and diglyceride contents were normalized to chow-fed levels in HF-fed itga1(-/-) mice. This indicates that hepatic steatosis is dissociated from insulin resistance in HF-fed itga1(-/-) mice. The decrease in hepatic lipid accumulation in HF-fed itga1(-/-) mice was associated with altered free fatty acid metabolism. These studies establish a role for integrin signaling in facilitating hepatic insulin action while promoting lipid accumulation in mice challenged with a HF diet.


Subject(s)
Fatty Liver/metabolism , Glucose/metabolism , Insulin Resistance/genetics , Integrin alpha1/biosynthesis , Animals , Diet, High-Fat , Fatty Liver/pathology , Hepatocytes/metabolism , Humans , Insulin/metabolism , Integrin alpha1/genetics , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Triglycerides/metabolism
7.
J Neuroinflammation ; 10: 33, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23448258

ABSTRACT

BACKGROUND: There is increasing evidence to suggest that pericytes play a crucial role in regulating the remodeling state of blood vessels. As cerebral pericytes are embedded within the extracellular matrix (ECM) of the vascular basal lamina, it is important to understand how individual ECM components influence pericyte remodeling behavior, and how cytokines regulate these events. METHODS: The influence of different vascular ECM substrates on cerebral pericyte behavior was examined in assays of cell adhesion, migration, and proliferation. Pericyte expression of integrin receptors was examined by flow cytometry. The influence of cytokines on pericyte functions and integrin expression was also examined, and the role of specific integrins in mediating these effects was defined by function-blocking antibodies. Expression of pericyte integrins within remodeling cerebral blood vessels was analyzed using dual immunofluorescence (IF) of brain sections derived from the animal model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). RESULTS: Fibronectin and collagen I promoted pericyte proliferation and migration, but heparan sulfate proteoglycan (HSPG) had an inhibitory influence on pericyte behavior. Flow cytometry showed that cerebral pericytes express high levels of α5 integrin, and lower levels of α1, α2, and α6 integrins. The pro-inflammatory cytokine tumor necrosis factor (TNF)-α strongly promoted pericyte proliferation and migration, and concomitantly induced a switch in pericyte integrins, from α1 to α2 integrin, the opposite to the switch seen when pericytes differentiated. Inhibition studies showed that α2 integrin mediates pericyte adhesion to collagens, and significantly, function blockade of α2 integrin abrogated the pro-modeling influence of TNF-α. Dual-IF on brain tissue with the pericyte marker NG2 showed that while α1 integrin was expressed by pericytes in both stable and remodeling vessels, pericyte expression of α2 integrin was strongly induced in remodeling vessels in EAE brain. CONCLUSIONS: Our results suggest a model in which ECM constituents exert an important influence on pericyte remodeling status. In this model, HSPG restricts pericyte remodeling in stable vessels, but during inflammation, TNF-α triggers a switch in pericyte integrins from α1 to α2, thereby stimulating pericyte proliferation and migration on collagen. These results thus define a fundamental molecular mechanism in which TNF-α stimulates pericyte remodeling in an α2 integrin-dependent manner.


Subject(s)
Cerebrum/cytology , Cerebrum/metabolism , Integrin alpha1/biosynthesis , Integrin alpha2/biosynthesis , Pericytes/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Cerebrum/drug effects , Female , Mice , Mice, Inbred C57BL , Pericytes/drug effects , Tumor Necrosis Factor-alpha/physiology
8.
J Cell Mol Med ; 15(2): 196-208, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20015200

ABSTRACT

Tissue factor pathway inhibitor-2 (TFPI-2) is a potent inhibitor of plasmin which activates matrix metalloproteinases (MMPs) involved in degradation of the extracellular matrix. Its secretion in the tumour microenvironment makes TFPI-2 a potential inhibitor of tumour invasion and metastasis. As demonstrated in aggressive cancers, TFPI-2 is frequently down-regulated in cancer cells, but the mechanisms involved in the inhibition of tumour progression remained unclear. We showed in this study that stable TFPI-2 down-regulation in the National Cancer Institute (NCI)-H460 non-small cell lung cancer cell line using specific micro interfering micro-interfering RNA promoted tumour progression in a nude mice orthotopic model that resulted in an increase in cell invasion. Moreover, TFPI-2 down-regulation enhanced cell adhesion to collagen IV and laminin via an increase in α(1) integrin on cell surface, and increased MMP expression (mainly MMP-1 and -3) contributing to cancer cell invasion through basement membrane components. This study also reveals for the first time that pulmonary fibroblasts incubated with conditioned media from TFPI-2 silencing cancer cells exhibited increased expression of MMPs, particularly MMP-1, -3 and -7, that are likely involved in lung cancer cell invasion through the surrounding stromal tissue, thus enhancing formation of metastases.


Subject(s)
Carcinoma, Non-Small-Cell Lung/physiopathology , Glycoproteins/genetics , Glycoproteins/metabolism , Lung Neoplasms/physiopathology , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Collagen Type IV/metabolism , Down-Regulation , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha1/biosynthesis , Laminin/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Matrix Metalloproteinases/biosynthesis , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , RNA Interference , Signal Transduction , Stromal Cells , Transplantation, Heterologous , Tumor Microenvironment
9.
J Reprod Immunol ; 87(1-2): 39-44, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20688399

ABSTRACT

Although the female genital tract is the main portal of entry for sexually transmitted infections in women, we still have limited understanding of the generation, maintenance and characteristics of memory T cells in the local tissue. Here, we utilized a mouse model of intravaginal HSV-2 infection and tetramers against the immunodominant HSV glycoprotein B epitope recognized by CD8+ T cells to examine the generation, maintenance and characteristics of anti-HSV memory T cells in the genital tract following acute infection. Our results show that the highest percentage of HSVgB-specific CD8+ T cells was found in the genital tract compared to the spleen or iliac lymphnode. Indeed, although the actual number of CD8+ T cells contracted following viral clearance, approximately one quarter of the CD8+ population that remained in the genital tissue was HSVgB-specific. Memory gB-tetramer+CD8 T cells in the genital tract were positive for CD127 and KLRG1 and negative for CD62L and CCR7, thus confirming that HSV-specific CD8 cells were effector memory T cells that lack the capacity for homing to lymphoid tissues. Functionally, both memory CD8+ and CD4+ HSV-specific populations in the genital tract produced IFNγ when stimulated in vitro and CD4+ cells also produced TNFα. Genital HSVgB-specific memory T cells expressed tissue-homing integrins CD103 (αE integrin) and CD49a (VLA-1 or α1 integrin). Our findings suggest that HSV-specific memory T cells are retained in the genital tract, poised to act as an early line of defense against future virus encounter.


Subject(s)
Genitalia, Female/immunology , Herpes Genitalis/immunology , Herpesvirus 2, Human/immunology , Immunologic Memory , T-Lymphocytes/immunology , Viral Envelope Proteins/immunology , Animals , Antigens, CD/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Female , Flow Cytometry , Genitalia, Female/virology , Herpes Genitalis/virology , Integrin alpha Chains/biosynthesis , Integrin alpha1/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-7 Receptor alpha Subunit/metabolism , Lectins, C-Type/metabolism , Mice , Mice, Inbred C57BL , Receptors, Immunologic , Spleen/immunology , T-Lymphocytes/metabolism , Trans-Activators/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
10.
Int J Oncol ; 33(4): 815-21, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18813796

ABSTRACT

Neuroblastoma is a childhood tumor thought to arise through improper differentiation of neural crest cells. MYCN amplification is a prognostic factor that indicates a highly malignant disease and poor patient prognosis. Integrins are important regulators of neuroblastoma attachment and migration and participate in many aspects of metastasis. However, the role of integrins in neuroblastoma metastasis, the leading cause of death from this disease, remains less well understood. Screening of neuroblastoma cell lines for integrin mRNA expression showed that integrin alpha1 expression was higher in lines such as SK-N-SH and NB69 that do not have MYCN amplification than in cell lines such as IMR32, NB1, NB9 and NB19 that have MYCN amplification. A knockdown of MYCN in NB1 and NB19 cells resulted in increased expression of integrin alpha1, which correlated with enhanced attachment to the extracellular matrix and reduced migratory activity. In contrast, the overexpression of MYCN in SK-N-SH and NB69 cells resulted in decreased expression of integrin alpha1, which correlated with reduced attachment to the extracellular matrix and enhanced migratory activity. These results show that MYCN may limit cell adhesion to the extracellular matrix and promote cell migration by downregulating integrin alpha1.


Subject(s)
Brain Neoplasms/metabolism , Down-Regulation , Genes, myc , Integrin alpha1/biosynthesis , Neuroblastoma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Humans , Integrin alpha1/genetics , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Neuroblastoma/pathology , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , RNA, Small Interfering/metabolism
11.
Acta Histochem ; 110(5): 363-70, 2008.
Article in English | MEDLINE | ID: mdl-18377961

ABSTRACT

Irregular or low expression of integrins, which are cell adhesion molecules, may be associated with infertility. We conducted a prospective controlled study evaluating the effects of supraphysiological levels of estrogen and progesterone created by human menopausal gonadotropins (HMG) and progesterone support on alpha(1)-integrin immunolocalisation in the endometrium. Three groups were enrolled in the study. The first group of patients (group 1) had unexplained infertility and had been treated with HMG and progesterone (n=27). The second group of patients (group 2) was an untreated fertile group (n=24). The third group (group 3) consisted of patients who had unexplained infertility and had received no treatment (n=11). Endometrial biopsy specimens were taken from individuals from each group during the ovulation induction period. alpha(1)-integrin immunohistochemistry was performed. Serum estradiol and progesterone levels were also measured in parallel with histological dating of endometrial biopsies. Group 1 showed no statistical difference from group 2 in alpha(1)-integrin or histological dating. Group 3 showed less alpha(1)-integrin in the glandular epithelium in the secretory phase. We observed that alpha(1)-integrin was specific to the secretory phase. Its localization was denser in group 2 when compared with group 3, which supports the conclusion that alpha(1)-integrin may be a useful marker for luteal phase quality. Moreover, the supraphysiological estrogen and progesterone levels created by HMG and progesterone support may affect the alpha(1)-integrin in the endometrium in the secretory phase in the case of unexplained infertile patients.


Subject(s)
Endometrium/metabolism , Infertility/metabolism , Integrin alpha1/biosynthesis , Progesterone/metabolism , Adult , Biomarkers , Biopsy , Body Mass Index , Estrogens/biosynthesis , Female , Gonadotropins/metabolism , Humans , Immunohistochemistry/methods , Infertility/etiology , Progesterone/biosynthesis , Prospective Studies , Time Factors
12.
Zhonghua Yi Xue Za Zhi ; 88(2): 119-22, 2008 Jan 08.
Article in Chinese | MEDLINE | ID: mdl-18353220

ABSTRACT

OBJECTIVE: To study the mechanism of migration of hepatic stellate cells (HSCs) within the space of Disse microenvironment during liver fibrosis, and to explore the novel pathogenesis of liver fibrosis from the view of cell migration. METHODS: Human HSCs of the line LX2-HSC were cultured. A modified in vitro Boyden chamber system was used to partially mimic the microenvironment of Disse space of normal basement membrane like matrix or that in fibrosis. HSCs were put in the upper chamber, and transforming growth factor (TGF)-beta1, plate-derived growth factor (PDGF)-BB, epidermal growth factor (EGF), vascular epithelial growth factor (VEGF), bfibroblast growth factor (bFGF), and collagen type I or type IV were put into the lower chamber. Four hours later cell migration assay was conducted. HSCs were put into 6-well plate and then added with TGF-beta1, PDGF-BB, EGF, VEGF, bFGF, and collagen type I or type IV, zymography was used to examine the activity of matrix metalloproteinases 2 and 9. SDS-gel immunoblotting was used too. RESULTS: Stimulation of HSCs with PDGF-BB, TGF-beta1, and/or EGF resulted in an increase in their migratory capacity and up-regulated MMP-2 activity. And the increase of MMP-2 could enhance Migration of HSC by 4.9-fold. The migration of HSCs (3.2-fold) was induced by type I collagen and inhibited by type IV collagen (1.2-fold). Migration induced by PDGF-BB, TGF-beta1, and collagen I could be inhibited by alpha1- and/or alpha2-integrin blocking antibodies. CONCLUSION: In liver fibrosis, alterations within the space of Disse microenvironment facilitate the migration of HSCs; the mechanism is associated with up-regulation of MMP-2 and with mediation of alpha1beta1 and alpha2beta1 integrins. Extracellular matrix by it self shows feedback actions to migration of HSCs.


Subject(s)
Cell Movement , Liver Cirrhosis/physiopathology , Cells, Cultured , Extracellular Matrix/metabolism , Humans , Integrin alpha1/biosynthesis , Integrin alpha2/biosynthesis , Liver/metabolism , Liver/pathology , Liver Cirrhosis/pathology , Matrix Metalloproteinase 2/biosynthesis
13.
J Immunol ; 179(8): 5054-63, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17911590

ABSTRACT

Primary viral infections of the lung induce potent effector CD8 T cell responses. To function in the influenza-infected airways, CD8 T cells must be able to resist cell death. The majority of the CD8 T cells in the airways and lung parenchyma expressed CD49a, the alpha-chain of the type IV collagen receptor VLA-1, and these cells were highly activated, producing both IFN-gamma and TNF-alpha. In the airways, where type IV collagen is abundant, but not the spleen, the CD49a(+) CD8 cells had reduced proportions of annexin V and caspase 8, and >80% expressed the TNF-alpha receptor II, while Fas, TNFR-I, and CD27 expression were similar to CD49a(-) cells. Furthermore, the CD49a(+), but not CD49a(-), CD8 T cells from the airways were resistant to active induction of apoptosis in the presence of type IV collagen and TNF-alpha in vitro. We propose that TNFR-II and the VLA-1 synergize to protect effector CD8 T cells in the infected airways from apoptosis during the acute infection.


Subject(s)
Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Integrin alpha1beta1/physiology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Receptors, Tumor Necrosis Factor, Type II/physiology , Respiratory Mucosa/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Separation , Female , Immunophenotyping , Influenza A Virus, H3N2 Subtype/immunology , Integrin alpha1/biosynthesis , Integrin alpha1/metabolism , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/pathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
14.
J Clin Immunol ; 27(6): 580-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17891451

ABSTRACT

The alpha1beta1 integrin, very late antigen (VLA)-1, characterizes collagen adherent interferon (IFN) gamma producing memory T cells in inflamed synovium. We now report that the mean percentage of VLA-1+ T cells is significantly lower among peripheral blood mononuclear cells of rheumatoid patients responsive to antitumor necrosis factor (TNF) alpha therapy than of those with active disease not receiving therapy. Neutralization of TNFalpha during in vitro polyclonal activation of VLA-1- T cells reduced differentiation to expression of VLA-1 and inhibited secretion of IFNgamma, but did not affect integrin expression on in vivo differentiated VLA-1+ T cells. Moreover, synovial fluids of patients relapsing during and after therapy were enriched in VLA-1+ T cells and lines derived from VLA-1+ T cells in peripheral blood of treated patients retained collagen binding and secreted IFN gamma. Thus, whereas therapy decreases VLA-1+ T cells in rheumatoid arthritis patients, a subset is resistant and contributes to residual and recurring inflammation.


Subject(s)
Arthritis, Rheumatoid/immunology , Integrin alpha1/biosynthesis , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Aged , Antibodies, Blocking/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/therapy , Cell Line , Down-Regulation/immunology , Female , Humans , Infliximab , Integrin alpha1/physiology , Lymphocyte Activation/immunology , Male , Middle Aged , Recurrence , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/immunology
15.
Biochim Biophys Acta ; 1769(9-10): 546-58, 2007.
Article in English | MEDLINE | ID: mdl-17669516

ABSTRACT

The integrin collagen receptor locus on human chromosome 5q11.2 includes the integrin genes ITGA1 and ITGA2, and the cell cycle regulation gene PELO, embedded within ITGA1 intron 1. ITGA1 contains a CArG box that is bound by serum response factor (SRF), while PELO contains two Sp1 binding elements. A comparison of mRNA levels in megakaryocytic (MK) and non-megakaryocytic (non-MK) cell lines and an analysis of the transcriptional activity of promoter-LUC reporter gene constructs in transfected cells revealed that ITGA1 is selectively suppressed in the MK lineage. Sodium bisulfite genomic sequencing established that a CpG-rich ITGA1 promoter region (-209/+115) is fully methylated at 19 CpG sites in MK cells that do not express alpha1beta1, but completely demethylated in expressing cells. In vitro methylation of ITGA1 suppresses transcription, while treatment of megakaryocytic cells with 5-aza-2'-deoxycytidine, but not Trichostatin A, resulted in de novo expression of ITGA1. During thrombopoietin-induced in vitro differentiation of primary human cord blood mononuclear cells into megakaryocytes, we observed rapid, progressive CpG methylation of ITGA1, but not PELO or ITGA2. Thus, selective CpG methylation of the ITGA1 promoter is a specific feature of alpha1beta1 regulation that coincides with the initiation of megakaryocyte differentiation.


Subject(s)
Chromosomes, Human, Pair 5/metabolism , DNA Methylation , Epigenesis, Genetic/physiology , Integrin alpha1/biosynthesis , Integrin alpha1beta1/biosynthesis , Integrin alpha2/biosynthesis , Megakaryocytes/metabolism , Nuclear Proteins/biosynthesis , Quantitative Trait Loci/physiology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Chromosomes, Human, Pair 5/genetics , DNA Methylation/drug effects , Decitabine , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , HeLa Cells , Humans , Hydroxamic Acids/pharmacology , Integrin alpha1/genetics , Integrin alpha1beta1/genetics , Integrin alpha2/genetics , Introns/physiology , K562 Cells , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Megakaryocytes/cytology , Nuclear Proteins/genetics , Promoter Regions, Genetic/physiology , Thrombopoietin/pharmacology , Transcription, Genetic
16.
Melanoma Res ; 17(4): 215-23, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17625451

ABSTRACT

Expression of collagen receptor integrins alpha1beta1 and alpha2beta1 has been associated with progression and metastatic potential of malignant melanoma. Integrin alpha2beta1 was originally characterized as a melanoma progression antigen. We have used real-time quantitative PCR to study the mRNA expression levels of three collagen receptor integrin chains, that is alpha1, alpha2 and alpha11 in metastases from 26 patients with melanoma. Interestingly, we find that survival after initiation of chemoimmunotherapy was significantly decreased in all patients whose tumours expressed high mRNA levels of alpha1 integrin, alpha2 integrin or alpha11 integrin when compared with lower tumour expression levels (P<0.05, log rank test). Moreover, those patients with high mRNA levels of all studied integrins had a significantly shorter survival from the appearance of the first metastasis than the patients with low levels of integrins (P<0.05). Furthermore, a high mRNA expression level of integrin alpha2 was found to be associated with poorer overall survival. High alpha2 mRNA levels (n=6) were associated with median survival of 35 months and low alpha2 mRNA levels (n=20), with median survival of 53 months (P=0.033). We conclude that collagen receptor integrins are important in the progression and prognosis of metastatic melanoma, and their measurements might be used as predictive markers when assessing disease progression.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Melanoma/mortality , Skin Neoplasms/metabolism , Skin Neoplasms/mortality , Adult , Aged , Disease-Free Survival , Female , Humans , Integrin alpha1/biosynthesis , Integrin alpha2/biosynthesis , Male , Melanoma/drug therapy , Middle Aged , Models, Biological , Receptors, Collagen/metabolism , Skin Neoplasms/drug therapy
17.
Spine (Phila Pa 1976) ; 31(7): 736-41, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16582846

ABSTRACT

STUDY DESIGN: In vitro investigation of vertebral endplate chondrocyte apoptosis. OBJECTIVES: To determine whether Fas antibody caused apoptosis in endplate chondrocytes, and whether insulin-like growth factor-1 (IGF-1) inhibited this effect. Integrin-alpha1 and focal adhesion kinase (FAK) expression in conjunction with apoptosis was also investigated. SUMMARY OF BACKGROUND DATA: Binding of Fas antibody to Fas mimics Fas-FasL ligation, which causes apoptosis. IGF-1 has been shown to have anti-apoptotic effects. MATERIALS AND METHODS: Rat cervical endplate chondrocytes were cultured and treated with Fas antibody, with or without IGF-1. Cellular morphology was examined by microscopy. Apoptotic changes were evaluated by transmission electron microscopy, TUNEL staining, and immunostaining. Apoptosis-induced changes in the expression of integrin-alpha1 chain and FAK were also investigated. RESULTS: Endplate chondrocytes were able to be cultured; a chondrocytic phenotype was maintained. Fas antibody induced apoptosis in endplate chondrocytes; this was confirmed by TUNEL staining. Bcl-2 expression was decreased by Fas antibody, while Bax expression increased. Integrin-alpha1 and FAK expression was decreased by Fas antibody. IGF-1 treatment inhibited these Fas antibody-induced changes. CONCLUSIONS: Fas antibody induces apoptosis and decreases Integrin-alpha1 and FAK expression in cultured endplate chondrocytes; IGF-1 is protective against these changes.


Subject(s)
Antibodies, Monoclonal/physiology , Apoptosis/physiology , Chondrocytes/metabolism , Insulin-Like Growth Factor I/pharmacology , fas Receptor/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Murine-Derived , Antibody Affinity , Apoptosis/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Fas Ligand Protein , Integrin alpha1/biosynthesis , Membrane Glycoproteins/metabolism , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factors/metabolism , fas Receptor/metabolism
18.
Front Biosci ; 11: 1302-10, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16368517

ABSTRACT

Stem cells responsible for maintenance and repair of tissues are found in a number of organs. The liver's remarkable capacity to regenerate after hepatectomy or chemical-induced injury does not involve proliferation of stem cells. However, recent studies suggest that liver stem cells exist in both embryonic and adult livers. Using fluorescence-activated cell sorting and a culture system in which primitive hepatic progenitor cells form colonies, a novel class of cells with the marker profile c-Met(+)CD49f(+/low)c-Kit(-)CD45(-)TER119(-) was found in the developing liver. This class apparently represents the population of cells that form colonies containing distinct hepatocytes and cholangiocytes. When cells in this class are transplanted into the spleen or liver of mice subjected to liver injury, the cells migrate and differentiate into liver parenchymal cells and cholangiocytes that are morphologically and functionally indistinguishable from their native counterparts. During mid-gestation, hematopoietic cells migrate into the liver from a region bounded by aorta, gonad, and mesonephros and produce oncostatin M (OSM). In combination with glucocorticoid hormones, OSM induces maturation of liver stem and progenitor cells, including those of the c-Met(+)CD49f(+/low)c-Kit(-)CD45(-)TER119(-) class. The ability to manipulate the proliferation and differentiation of liver stem cells in vitro will greatly aid in analyzing mechanisms of liver development and offers promise in stem cell therapy of liver diseases.


Subject(s)
Gene Expression Regulation, Developmental , Liver/cytology , Liver/embryology , Stem Cells/pathology , Animals , Cell Proliferation , Cell Separation , Cell Transplantation , Cytokines/metabolism , Extracellular Matrix/metabolism , Flow Cytometry , Glycophorins/biosynthesis , Hematopoietic Stem Cells/metabolism , Hepatocyte Nuclear Factors/metabolism , Hepatocytes/metabolism , Humans , Integrin alpha1/biosynthesis , Leukocyte Common Antigens/biosynthesis , Liver/metabolism , Liver/pathology , Mice , Models, Biological , Oncostatin M , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/metabolism
19.
Oncol Rep ; 14(4): 941-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16142355

ABSTRACT

To investigate the interaction between anticancer drug resistance and radioresistance in cervical cancer cells, 3 single cell-derived cyclophosphamide-resistant subclones were established from the drug- and radiosensitive human cervical squamous cell carcinoma cell line ME180 by chronic exposure cultures with 4-hydroxy-cyclophosphamide followed by limiting dilution. The established cyclophosphamide-resistant subclones were also radio- and multidrug-resistant to 7 other anticancer drugs. Flow cytometric analysis revealed significantly increased levels of CD40 expression on the 3 resistant subclones, whereas no CD40 expression was found on the parent ME180 cells. However, there were no changes in the expression levels of CD29, CD49a-CD49f or CD59 between the parent cells and resistant subclones. A recombinant human soluble CD40 ligand had no effect on the proliferation of the resistant subclones. Irradiation had no effect on the 4-hydroxy-cyclophosphamide sensitivity of the parent cells. These results indicate that the established cyclophosphamide-resistant subclones have impaired cell death signals, which are common to both drug- and radiation-induced apoptosis, and cyclophosphamide may not be an adequate drug for use in concurrent chemoradiotherapy. Furthermore, CD40 activation signals may be associated with the multidrug- and radioresistance in these cyclophosphamide-resistant subclones.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , CD40 Antigens/biosynthesis , Carcinoma, Squamous Cell/drug therapy , Cyclophosphamide/therapeutic use , Drug Resistance, Neoplasm , Uterine Cervical Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Bleomycin/pharmacology , CD40 Ligand/metabolism , CD59 Antigens/biosynthesis , Cell Line, Tumor , Cell Proliferation , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/metabolism , Cyclophosphamide/pharmacology , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Resistance, Multiple , Female , Flow Cytometry , Gamma Rays , Humans , Integrin alpha1/biosynthesis , Integrin alpha2/biosynthesis , Integrin alpha3/biosynthesis , Integrin alpha4/biosynthesis , Integrin alpha5/biosynthesis , Integrin alpha6/biosynthesis , Integrin beta1/biosynthesis , Radiation Tolerance
20.
Am J Physiol Gastrointest Liver Physiol ; 288(1): G67-73, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15319185

ABSTRACT

Insulitis is a hallmark feature of autoimmune diabetes that ultimately results in islet beta-cell destruction. We examined integrin requirements and specific inhibition of integrin structure in T cell and monocyte adhesion to pancreatic islet endothelium. Examination of cell surface integrin expression on WEHI 7.1 T cells revealed prominent expression of beta-, beta(1)-, alpha(L)-integrins, and low expression of alpha(M)-integrins; whereas WEHI 274.1 monocytes showed significant staining for beta(2)-, beta(1)-, alpha(M)-molecules and no expression of alpha(L)-molecules. Unstimulated islet endothelium showed constitutive levels of ICAM-1 counter-ligand expression with minimal VCAM-1 expression; however, TNF-alpha stimulation increased cell surface density of both molecules. TNF-alpha increased T cell and monocyte rolling and adhesion under hydrodynamic flow conditions. Administration of a cyclic peptide competitor for the alpha(L)-integrin I domain binding sites (cyclo1,12-PenITDGEATDSGC) blocked T cell adhesion without inhibiting monocyte adhesion. Examination of T cell rolling revealed that cLAB.L treatment increased the average rolling velocity on activated endothelium and significantly decreased the fraction of T cells rolling at < or =50 microm/s, suggesting that cLAB.L treatment interferes with signal activation events required for the conversion of T cell rolling to firm adhesion. These data demonstrate for the first time that cyclic peptide antagonists against alpha(L)-integrin I domain attenuate T cell recruitment to islet endothelium.


Subject(s)
Cell Adhesion , Diabetes Mellitus, Type 1/physiopathology , Integrin alpha1/biosynthesis , Integrin alpha1/pharmacology , Islets of Langerhans/physiology , T-Lymphocytes/physiology , Animals , Integrins/biosynthesis , Integrins/physiology , Mice , Monocytes/physiology , Peptide Fragments , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...