Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Reprod Sci ; 31(4): 1159-1169, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38097900

ABSTRACT

The efficacy of human amniotic mesenchymal stem cell (hAMSC) ovarian injection in improving ovarian function in primary ovarian insufficiency (POI) patients has been shown in some reports. However, the safety and efficacy of hAMSC vein injection remains unclear. In this study, we evaluated the safety and efficacy of hAMSC intravenous injection in cynomolgus macaques and SD rats and provided evidence for clinical trials. The hAMSCs were transplanted three times in SD rats at low, medium, and high doses. The animal behavior and biochemical and biophysical parameters were routinely monitored on a 2-month period posttransplantation, and histopathologic examinations were also performed. Experiments on the acute toxicity, allergy test, and hemolysis test showed that hAMSCs possess good biocompatibility. Our results showed that the maximum tolerated dose of hAMSCs in SD rats was 4.0 × 107 cells/kg. The maximum safe dose with three injections of hAMSCs in SD rats was 5.0 × 106 cells/kg. In addition, the results demonstrated that hAMSCs may restore POI rat ovarian function after two injections of 2.5 × 106 cells/kg or 5.0 × 106 cells/kg, which improved the disturbed estrous cycle, hormone levels, and ovarian lesions induced by pZP3. In conclusion, the preclinical results suggested that the transplantation of hAMSCs may be safe and efficacious for SD rats at doses of 5.0 × 106 cells/kg and lower.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Ovarian Cysts , Ovarian Neoplasms , Primary Ovarian Insufficiency , Female , Humans , Rats , Animals , Primary Ovarian Insufficiency/metabolism , Ovarian Cysts/metabolism , Rats, Sprague-Dawley , Mesenchymal Stem Cell Transplantation/methods , Ovarian Neoplasms/metabolism , Mesenchymal Stem Cells/metabolism
2.
iScience ; 26(7): 107057, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37534158

ABSTRACT

Bipolar disorder (BD) is a common mental disorder characterized by manic and depressive episodes. Mood disorders have been associated with immune dysfunction. The combination of quetiapine and valproate has shown positive effects in treating BD, but the impact on immune dynamics remains less understood. Using single-cell RNA sequencing, we observed that B cells exhibited downregulation of inflammation-related genes, while pro-inflammatory mast and eosinophil cells decreased following treatment. Ribosomal peptide production genes were found to be reduced in both B and T cells after treatment. Additionally, our findings suggest that the combined therapy effectively alleviates inflammation by reducing myloid-mediated immune signaling pathways. This study provides valuable insights into the immune atlas and uncovers a potential mechanism for immune disorder alleviation in patients with BD treated with quetiapine and valproate.

3.
Front Pharmacol ; 14: 1067665, 2023.
Article in English | MEDLINE | ID: mdl-37033662

ABSTRACT

Nav1.5 channel is crucial for the proliferation and migration of breast cancer cells. In this study, we investigated the anticancer effect of JZTX-14, a natural peptide considered an effective antagonist of Nav1.5. First, we successfully isolated and purified the 31 amino acid peptide JZTX-14 containing three pairs of disulfide bonds from spider venom and synthesised JZTX-14 by solid phase synthesis. We then predicted their physiochemical properties and structures in the peptide database. Further, we investigated the effects of natural and synthetic JZTX-14 on the proliferation and migration of MDA-MB-231 breast cancer cells via modulation of sodium current through the Nav1.5 channel. The results showed that both synthetic and natural JZTX-14 inhibited Nav1.5 currents, indicating the successful synthesis of JZTX-14. However, JZTX-14 did not affect MDA-MB-231 cell proliferation but inhibited its migration. Transcriptome analysis revealed that JZTX-14 downregulated S100A4 and FBXO2 and upregulated SERPINB2 in MDA-MB-231 cells. Western blot analysis demonstrated an increased level of the epithelial marker, E-cadherin, and decreased levels of the mesenchymal markers, N-cadherin and vimentin, and matrix metalloproteinase (MMP2), indicating the possible underlying mechanism of the inhibition of MDA-MB-231 cell migration by JZTX-14. This study provides a new target for inhibiting breast cancer metastasis and identifies a potent natural peptide for treating Triple-negative breast cancer.

4.
J Cell Physiol ; 234(7): 10855-10867, 2019 07.
Article in English | MEDLINE | ID: mdl-30536377

ABSTRACT

The key signaling networks regulating bone marrow mesenchymal stem cells (BMSCs) are poorly defined. Lgr4, which belongs to the leucine-rich repeat-containing G protein-coupled receptor (LGR) family, is widely expressed in multiple tissues from early embryogenesis to adulthood. We investigated whether Lgr4 functions in BMSCs and in osteogenesis, adipogenesis, and skeletal myoblasts, using mice with a ß-geo gene trap inserted into the Lgr4 gene. Abundant Lgr4 expression was detected in skeletal, adipose and muscular tissue of Lgr4+/- mice at E16.5 by ß-gal staining, and Lgr4-deficiency promoted BMSC proliferation (16 ± 4 in wild-type [WT] and 28 ± 2 in Lgr4-/- ) using colony forming units-fibroblast assay, while suppressing BMSC migration (from 103 ± 18 in WT to 57 ± 10 in Lgr4-/- ) by transwell migration assay and apoptosis ratio (from 0.0720 ± 0.0123 to 0.0189 ± 0.0051) by annexin V staining assay. Deletion of Lgr4 decreased bone mass (BV/TV from 19.16 ± 2.14 in WT mice to 10.36 ± 1.96 in KO) and fat mass through inhibiting BMSC differentiation to osteoblasts or adipocytes. Furthermore, LGR4-regulated osteogenic, adipogenic, and myogenic gene expression. Importantly, our data showed that loss of Lgr4-inhibited fracture healing by suppressing osteoblast differentiation. Moreover, deletion of Lgr4 in BMSCs-delayed fracture healing following stem cell therapy by BMSC transplantation. Together, our results demonstrated that LGR4 is essential for mesoderm-derived tissue development and BMSC differentiation, demonstrating that LGR4 could be a promising drug target for related diseases and a critical protein for stem cell therapy.


Subject(s)
Apoptosis , Cell Differentiation , Cell Movement , Cell Proliferation , Femur/metabolism , Mesenchymal Stem Cells/metabolism , Receptors, G-Protein-Coupled/deficiency , Tibia/metabolism , Adipocytes/metabolism , Adipogenesis , Animals , Femur/cytology , Gestational Age , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Osteoblasts/metabolism , Osteogenesis , Phenotype , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Tibia/cytology
5.
Nat Med ; 22(5): 539-46, 2016 05.
Article in English | MEDLINE | ID: mdl-27064449

ABSTRACT

Tumor necrosis factor (TNF) superfamily member 11 (TNFSF11, also known as RANKL) regulates multiple physiological or pathological functions, including osteoclast differentiation and osteoporosis. TNFRSF11A (also called RANK) is considered to be the sole receptor for RANKL. Herein we report that leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4, also called GPR48) is another receptor for RANKL. LGR4 competes with RANK to bind RANKL and suppresses canonical RANK signaling during osteoclast differentiation. RANKL binding to LGR4 activates the Gαq and GSK3-ß signaling pathway, an action that suppresses the expression and activity of nuclear factor of activated T cells, cytoplasmic, calcineurin-dependent 1 (NFATC1) during osteoclastogenesis. Both whole-body (Lgr4(-/-)) and monocyte conditional knockout mice of Lgr4 (Lgr4 CKO) exhibit osteoclast hyperactivation (including elevation of osteoclast number, surface area, and size) and increased bone erosion. The soluble LGR4 extracellular domain (ECD) binds RANKL and inhibits osteoclast differentiation in vivo. Moreover, LGR4-ECD therapeutically abrogated RANKL-induced bone loss in three mouse models of osteoporosis. Therefore, LGR4 acts as a second RANKL receptor that negatively regulates osteoclast differentiation and bone resorption.


Subject(s)
Bone Remodeling/genetics , Bone Resorption/genetics , Osteogenesis/genetics , RANK Ligand/metabolism , Receptors, G-Protein-Coupled/genetics , Animals , Blotting, Western , Calcium/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Giant Cell Tumor of Bone/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Leukocytes, Mononuclear , Mice , Mice, Knockout , Molecular Docking Simulation , NFATC Transcription Factors/metabolism , Optical Imaging , Osteoporosis/genetics , Osteoporosis/metabolism , RAW 264.7 Cells , Real-Time Polymerase Chain Reaction , Signal Transduction , Surface Plasmon Resonance , X-Ray Microtomography
6.
Sci Rep ; 5: 17605, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26620037

ABSTRACT

Excessive RANKL signaling leads to superfluous osteoclast formation and bone resorption, is widespread in the pathologic bone loss and destruction. Therefore, targeting RANKL or its signaling pathway has been a promising and successful strategy for this osteoclast-related diseases. In this study, we examined the effects of xanthohumol (XN), an abundant prenylflavonoid from hops plant, on osteoclastogenesis, osteoclast resorption, and RANKL-induced signaling pathway using both in vitro and in vivo assay systems. In mouse and human, XN inhibited osteoclast differentiation and osteoclast formation at the early stage. Furthermore, XN inhibited osteoclast actin-ring formation and bone resorption in a dose-dependent manner. In ovariectomized-induced bone loss mouse model and RANKL-injection-induced bone resorption model, we found that administration of XN markedly inhibited bone loss and resorption by suppressing osteoclast activity. At the molecular level, XN disrupted the association of RANK and TRAF6, resulted in the inhibition of NF-κB and Ca(2+)/NFATc1 signaling pathway during osteoclastogenesis. As a results, XN suppressed the expression of osteoclastogenesis-related marker genes, including CtsK, Nfatc1, Trap, Ctr. Therefore, our data demonstrated that XN inhibits osteoclastogenesis and bone resorption through RANK/TRAF6 signaling pathways. XN could be a promising drug candidate in the treatment of osteoclast-related diseases such as postmenopausal osteoporosis.


Subject(s)
Bone Resorption/metabolism , Cell Differentiation/drug effects , Flavonoids/pharmacology , Humulus/chemistry , Osteoclasts/metabolism , Propiophenones/pharmacology , Animals , Bone Resorption/pathology , Dose-Response Relationship, Drug , Flavonoids/chemistry , Humans , Mice , Osteoclasts/pathology , Propiophenones/chemistry , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , TNF Receptor-Associated Factor 6/metabolism
7.
PLoS One ; 10(7): e0134051, 2015.
Article in English | MEDLINE | ID: mdl-26208278

ABSTRACT

The G protein-coupled receptor TGR5 is a membrane receptor for bile acids. Its agonism increases energy expenditure and controls blood glucose through secretion of glucagon-like peptide-1 in enteroendocrine cells. In this study, we explored the therapeutic potential of WB403, a small compound activating TGR5 which was identified by combining TGR5 targeted luciferase assay and active GLP-1 assay, in treating type 2 diabetes. After confirmation of TGR5 and GLP-1 stimulating activities in various cell systems, WB403 was examined in oral glucose tolerance test, and tested on different mouse models of type 2 diabetes for glycemic control and pancreatic ß-cell protection effect. As a result, WB403 exhibited a moderate TGR5 activation effect while promoting GLP-1 secretion efficiently. Interestingly, gallbladder filling effect, which was reported for some known TGR5 agonists, was not detected in this novel compound. In vivo results showed that WB403 significantly improved glucose tolerance and decreased fasting blood glucose, postprandial blood glucose and HbA1c in type 2 diabetic mice. Further analysis revealed that WB403 increased pancreatic ß-cells and restored the normal distribution pattern of α-cell and ß-cell in islets. These findings demonstrated that TGR5 activator WB403 effectively promoted GLP-1 release, improved hyperglycemia and preserved the mass and function of pancreatic ß-cells, whereas it did not show a significant side effect on gallbladder. It may represent a promising approach for future type 2 diabetes mellitus drug development.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose Intolerance/metabolism , Insulin-Secreting Cells/drug effects , Receptors, G-Protein-Coupled/agonists , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Glucagon/metabolism , Glucose Intolerance/pathology , Glucose Tolerance Test , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice , Postprandial Period
8.
J Bone Miner Res ; 27(6): 1298-1308, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22337253

ABSTRACT

Receptor activator of NF-κB ligand (RANKL) stimulation leads to the activation of mitogen-activated protein kinase (MAPK)/AP-1 and Ca2+­nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) signaling pathways in osteoclastogenesis. Targeting these pathways has been an encouraging strategy for bone-related diseases, such as postmenopausal osteoporosis. In this study, we examined the effects of caffeic acid 3,4-dihydroxy-phenethyl ester (CADPE) on osteoclastogenesis. In mouse bone marrow monocytes (BMMs) and RAW264.7 cells, CADPE suppressed RANKL-induced osteoclast differentiation and actin-ring formation in a dose-dependent manner within non­growth inhibitory concentrations at the early stage, while CADPE had no effect on macrophage colony-stimulating factor (M-CSF)-induced proliferation and differentiation. At the molecular level, CADPE inhibited RANKL-induced phosphorylation of MAPKs, including extracellular signal-regulated kinases 1/2 (ERK1/2), p38, and c-Jun N-terminal kinase (JNK), without significantly affecting the NF-κB signaling pathway. CADPE abrogated RANKL-induced activator protein 1 (AP-1)/FBJ murine osteosarcoma viral oncogene homolog (c-Fos) nuclear translocation and activation. Overexpression of c-Fos prevented the inhibition by CADPE of osteoclast differentiation. Furthermore, CADPE suppressed RANKL-induced the tumor necrosis factor receptor associated factor 6 (TRAF6) interaction with c-src tyrosine kinase (c-Src), blocked RANKL-induced the phosphorylation of protein kinase B (AKT), and inhibited RANKL-induced Ca2+ oscillation. As a result, CADPE decreased osteoclastogenesis-related marker gene expression, including NFATc1, TRAP, cathepsin K, and c-Src. To test the effects of CADPE on osteoclast activity in vivo, we showed that CADPE prevented ovariectomy-induced bone loss by inhibiting osteoclast activity. Together, our data demonstrate that CADPE suppresses osteoclastogenesis and bone loss through inhibiting RANKL-induced MAPKs and Ca2+-NFATc1 signaling pathways. CADPE is a novel agent in the treatment of osteoclast-related diseases, such as osteoporosis.


Subject(s)
Bone Resorption/prevention & control , Caffeic Acids/pharmacology , Mitogen-Activated Protein Kinases/metabolism , NFATC Transcription Factors/metabolism , Osteogenesis/drug effects , RANK Ligand/pharmacology , Transcription Factor AP-1/metabolism , Actins/metabolism , Animals , Biomarkers/metabolism , Bone Marrow Cells/pathology , Bone Resorption/enzymology , Bone Resorption/genetics , Bone Resorption/pathology , Calcium , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Female , Gene Expression Regulation/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-kappa B/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/enzymology , Osteoclasts/pathology , Osteogenesis/genetics , Ovariectomy
9.
J Bone Miner Res ; 26(3): 644-56, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20814972

ABSTRACT

Activation of NF-κB and MAPK/activator protein 1 (AP-1) signaling pathways by receptor activator NF-κB ligand (RANKL) is essential for osteoclast activity. Targeting NF-κB and MAPK/AP-1 signaling to modulate osteoclast activity has been a promising strategy for osteoclast-related diseases. In this study we examined the effects of maslinic acid (MA), a pentacyclic triterpene acid that is widely present in dietary plants, on RANKL-induced osteoclastogenesis, osteoclast function, and signaling pathways by in vitro and in vivo assay systems. In mouse bone marrow monocytes (BMMs) and RAW264.7 cells, MA inhibited RANKL-induced osteoclastogenesis in a dose-dependent manner within nongrowth inhibitory concentration, and MA decreased osteoclastogenesis-related marker gene expression, including TRACP, MMP9, c-Src, CTR, and cathepsin K. Specifically, MA suppressed osteoclastogenesis and actin ring formation at early stage. In ovariectomized mice, administration of MA prevented ovariectomy-induced bone loss by inhibiting osteoclast activity. At molecular levels, MA abrogated the phosphorylation of MAPKs and AP-1 activity, inhibited the IκBα phosphorylation and degradation, blocked NF-κB/p65 phosphorylation, nuclear translocation, and DNA-binding activity by downregulating RANK expression and blocking RANK interaction with TRAF6. Together our data demonstrate that MA suppresses RANKL-induced osteoclastogenesis through NF-κB and MAPK/AP-1 signaling pathways and that MA is a promising agent in the treatment of osteoclast-related diseases such as osteoporosis.


Subject(s)
Bone Resorption/prevention & control , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteoclasts/enzymology , Osteogenesis/drug effects , RANK Ligand/pharmacology , Triterpenes/pharmacology , Actins/metabolism , Animals , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Resorption/enzymology , Cell Differentiation/drug effects , Cell Line , Female , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/pathology , NFATC Transcription Factors/metabolism , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/pathology , Osteogenesis/genetics , Ovariectomy , Receptor Activator of Nuclear Factor-kappa B/metabolism , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor AP-1/metabolism
10.
Gene Regul Syst Bio ; 3: 115-29, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19838339

ABSTRACT

SG2NA is a member of the striatin protein family. In human and mouse, the SG2NA gene encodes two major protein isoforms: SG2NA alpha and SG2NA beta. The functions of these proteins, except for acting as the regulatory subunits for PP-2A, remain largely unknown. To explore the possible functions of SG2NA in lower vertebrates, we have isolated two SG2NA cDNAs from goldfish, Carassius auratus. Our results reveal that the first cDNA contains an ORF of 2118 bp encoding a deduced protein with 705 amino acids, and the second one 2148 bp coding for a deduced protein of 715 amino acids. Comparative analysis reveals that both isoforms belong to the alpha-type, and are named SG2NA alpha and SG2NA alpha(+). RT-PCR and western blot analysis reveal that the SG2NA gene is differentially expressed in 9 tissues examined. During goldfish development, while the SG2NA mRNAs remain relatively constant in the first 3 stages and then become decreased and fluctuated from gastrula to larval hatching, the SG2NA proteins are fluctuated, displaying a peak every 3 to 4 stages. Each later peak is higher than the earlier one and the protein expression level becomes maximal at hatching stage. Together, our results reveal that SG2NA may play an important role during goldfish development and also in homeostasis of most adult tissues.

11.
Sci China C Life Sci ; 52(8): 724-32, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19727590

ABSTRACT

It is well established that the protein serine/threonine phosphatase 2A (PP2A) plays very important roles in many different cellular processes, including cell proliferation and differentiation, gene expression, neurotransmission, apoptosis, and aging. PP2A consists of three heterogenic subunits: the scaffold subunit A, the catalytic subunit C, and the regulatory subunit B. While both the scaffold and the catalytic subunits contain only two forms, at least four families of the regulatory subunits, B, B', B'', and B''' have been identified. These regulatory subunits from different families are encoded by different genes and bear other functions besides directing the specificity of PP2A. To study the functions of the regulatory subunits of PP2A in lower vertebrates, we have cloned the full-length cDNA sequence of the gene encoding the regulatory subunit B'delta of PP2A from gold fish, Carassius auratus using 3'-RACE and 5'-RACE cloning strategies. Our results revealed that the full-length B'delta cDNA contains 2415 bp and encodes a protein of 555 amino acids. The B'delta protein displays a very high level of sequence identity with the B'delta regulatory subunit from other species of vertebrates. Regarding its expression pattern, RT-PCR revealed that the highest level of mRNA was detected in brain, a less level detected in liver, spermary, ovary, kidney and gill, and the lowest level detected in the fin. During different developmental stages of gold fish, the highest level of mRNA expression was detected at the stages of two-cell, multiple-cell, blastula and gastrula, and a decreased level of B'gamma mRNA was detected in other developmental stages. At the protein level, the highest expression level of B'delta protein was found in spermary, ovary, brain and heart, a less amount found in liver and the lowest level detected in kidney, gill and fin. Developmentally, B'delta protein was strongly expressed at the stages of two-cell, multiple-cell, blastula, gastrula, neurula, and optic vesicle, and then decreased at the stages of brain differentiation and eye pigmentation. These results suggest that B'delta appears to play a very important role during gold fish development and also in adult tissue homeostasis.


Subject(s)
Goldfish/genetics , Protein Phosphatase 2/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Consensus Sequence , DNA, Complementary/genetics , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Goldfish/growth & development , Goldfish/metabolism , Humans , Male , Mice , Molecular Sequence Data , Organ Specificity , Protein Subunits/genetics , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid
12.
Clin Lab ; 49(1-2): 65-9, 2003.
Article in English | MEDLINE | ID: mdl-12593477

ABSTRACT

OBJECTIVE: To explore the relationship between the RANTES, TGFbeta1 and amount of mast cells (MC) surrounding the implanted tumors. METHOD: Pieces of Walker 256 carcinosarcoma were implanted in the liver of 40 male Wistar rats and the formed intrahepatic implanted tumors were then divided into 3 groups: group without MC infiltration, group with little MC infiltration and group with MC infiltration; 8 normal rats served as control group. The sera of rats in the different groups were tested by ELISA to find the serum RANTES content of the tumor bearing rats, then the chemotactic activity of the serum RANTES of different tumor bearing groups vs peritoneal MC of normal rats was tested by the microBoyden chamber. RESULTS: The MC amount of the tumor bearing rats was quite different, some of them showed a significantly increased amount. The groups with more MC infiltration showed a higher RANTES content in the sera and a stronger chemotactic activity vs MC. CONCLUSION: The RANTES was an effective chemotactic factor to MC. The serum concentration of RANTES of the tumor bearing rats is related to the difference of the MC amount surrounding the tumor.


Subject(s)
Carcinoma 256, Walker/pathology , Chemokine CCL5/blood , Liver Neoplasms, Experimental/pathology , Mast Cells/pathology , Animals , Carcinoma 256, Walker/blood , Chemotaxis , Liver Neoplasms, Experimental/blood , Male , Rats , Rats, Wistar , Transforming Growth Factor beta/blood , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL
...