Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Arch Gynecol Obstet ; 304(3): 743-750, 2021 09.
Article in English | MEDLINE | ID: mdl-33547934

ABSTRACT

PURPOSE: To detect the methylation of Dickkopf-associated protein 1 (DKK-1) gene promoter in cervical exfoliated cells and to study its clinical significance in cervical squamous cell carcinoma (CSCC) and its relationship with high-risk HPV infection. METHODS: Methylation-specific PCR (MSP) was utilized to detect the methylation of DKK-1 gene promoter in cervical exfoliated cells from 40 patients with CSCC and 40 patients with chronic cervicitis in the Affiliated Hospital of Inner Mongolia Medical University. The methylation rate of DKK-1 gene promoter in different clinicopathological factors and its relationship with high-risk HPV infection was compared, and different detection methods were compared. RESULT: The degree of methylation of DKK-1 gene promoter in CSCC group was significantly higher than that in cervicitis group (P < 0.05). In CSCC group, the degree of methylation was significantly different in high-risk HPV infection, histological differentiation, tumor size, lymph node metastasis and the International Federation of Gynecology and Obstetrics (FIGO) staging (all P < 0.05). The degree of methylation is not related to the type of high-risk HPV infection (P > 0.05). The one-year survival rate of CSCC patients with high-risk HPV positive and DKK-1 gene promoter methylation is relatively low, only 74.1%. The sensitivity, specificity and accuracy of DKK-1 gene methylation combined with high-risk HPV detection were 96.7%, 78.0% and 85.0%, respectively. CONCLUSION: Methylation of DKK-1 gene promoter in cervical exfoliated cells of patients with CSCC is related to high-risk HPV infection and different clinicopathological factors, but the degree of methylation of DKK-1 gene is not related to the type of high-risk HPV infection. It may become an indicator different from HPV typing detection, which may play a shunt role in suggesting whether further invasive cervical examination is needed and reduce cervical invasive examination and overtreatment. It may be related to the survival rate of patients, which can be used to estimate the development and prognosis of CSCC and may play a good role in early warning in follow-up monitoring of CSCC after treatment. DKK1 gene methylation combined with HPV detection can improve the sensitivity, specificity and accuracy of diagnosis, which may improve the detection rate of early CSCC and make up for the deficiency of HPV and TCT detection. That may become a non-invasive screening method for CSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Intercellular Signaling Peptides and Proteins/genetics , Papillomavirus Infections/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervicitis/genetics , Adult , Aged , Base Sequence , DNA Methylation , Female , Humans , Methylation , Middle Aged , Papillomavirus Infections/complications , Polymerase Chain Reaction , Pregnancy
2.
Mol Med Rep ; 17(2): 3356-3363, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29257290

ABSTRACT

Parkinson's disease (PD) is one of the most common neurodegenerative diseases. For decades, the unilateral 6­hydroxydopamine (6­OHDA) rat model has been employed to investigate the pathogenesis and therapy of PD. However, the behavior and associated pathological features of the model long term have not previously been described dynamically. In the present study, the unilateral model was established by 6­OHDA injection in the striatum. The PD rat model was determined 2 weeks following surgery, according to the apomorphine (APO)­induced rotations, cylinder, rotarod and open field tests. TH­positive neurons and fibers in the substantia nigra pars compacta (SNpc) and striatum, respectively, and glial activation in the SNpc, determined by glial fibrillary acidic protein (GFAP) expression for astrocytes and CD11b (Mac1) expression for microglia, were detected by immunohistological staining. Correlation analysis was performed to understand the association between PD­associated behavior and pathology. The behavioral impairment progressively deteriorated during the process of experiment. In addition, the decrease in TH­positive neurons was associated with an increase in GFAP­ and Mac1­positive cells in the SNpc. Linear regression analysis indicated the association between behavioral and pathological changes. The results of the present study indicate that the APO­induced rotation, cylinder and rotarod tests are all sensitive and reliable strategies to predict the loss of TH+ neurons. These results provide a potential intervention time­point and a comprehensive evaluation index system for assessment of PD therapeutic strategies using the hemiparkinsonian rat.


Subject(s)
Hydroxydopamines , Parkinson Disease, Secondary/pathology , Animals , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Disease Models, Animal , Male , Motor Activity , Neurons/pathology , Parkinson Disease, Secondary/physiopathology , Rats , Rats, Sprague-Dawley , Rotarod Performance Test , Substantia Nigra/pathology , Substantia Nigra/physiopathology
3.
Zhong Yao Cai ; 37(1): 1-4, 2014 Jan.
Article in Chinese | MEDLINE | ID: mdl-25090694

ABSTRACT

OBJECTIVE: To explore the effects of different hormonal combinations on induction, proliferation and differentiation of Orostachyis fimbriatae callus culture. METHODS: Aseptic seedling leaves were used as explants,the different concentrations of IAA,NAA, 6-BA and KT on induction proliferation of callus were optimized by orthogonal test to explore the optimum medium for differentiation of callus by tissue culture techniques. RESULTS: The best medium for induction was MS + IAA 1.0 mg/L + NAA 0.5 mg/L + KT 1.0 mg/L, and the best hormonal combination for proliferation was MS + IAA 0.5 mg/L + 6-BA 0.5 mg/I. + KT 1.0 mg/L. The best medium for differentiation was MS + IAA 0.1 mg/L + KT 2.0 mg/L, and 1/2MS + IAA 0.2 mg/L was the optimum medium for rooting culture. CONCLUSION: The system of regeneration of Orostachyis fimbriatae is establishd by tissue culture techniques in this study.


Subject(s)
Crassulaceae/physiology , Plant Growth Regulators/pharmacology , Regeneration/drug effects , Tissue Culture Techniques/methods , Crassulaceae/drug effects , Culture Media/chemistry , Plant Leaves/drug effects , Plant Leaves/physiology , Seeds/drug effects , Seeds/physiology
4.
J Immunol ; 192(10): 4610-9, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24719464

ABSTRACT

Identifying cross-species similarities and differences in immune development and function is critical for maximizing the translational potential of animal models. Coexpression of CD21 and CD24 distinguishes transitional and mature B cell subsets in mice. In this study, we validate these markers for identifying analogous subsets in humans and use them to compare the nonmemory B cell pools in mice and humans, across tissues, and during fetal/neonatal and adult life. Among human CD19(+)IgM(+) B cells, the CD21/CD24 schema identifies distinct populations that correspond to transitional 1 (T1), transitional 2 (T2), follicular mature, and marginal zone subsets identified in mice. Markers specific to human B cell development validate the identity of marginal zone cells and the maturation status of human CD21/CD24 nonmemory B cell subsets. A comparison of the nonmemory B cell pools in bone marrow, blood, and spleen in mice and humans shows that transitional B cells comprise a much smaller fraction in adult humans than mice. T1 cells are a major contributor to the nonmemory B cell pool in mouse bone marrow, in which their frequency is more than twice that in humans. Conversely, in spleen, the T1:T2 ratio shows that T2 cells are proportionally ∼ 8-fold higher in humans than in mice. Despite the relatively small contribution of transitional B cells to the human nonmemory pool, the number of naive follicular mature cells produced per transitional B cell is 3- to 6-fold higher across tissues than in mice. These data suggest differing dynamics or mechanisms produce the nonmemory B cell compartments in mice and humans.


Subject(s)
Antigens, CD19/immunology , B-Lymphocytes/immunology , CD24 Antigen/immunology , Receptors, Complement 3d/immunology , Adult , Animals , B-Lymphocytes/cytology , Humans , Infant, Newborn , Male , Mice , Middle Aged , Species Specificity
5.
PLoS One ; 8(5): e64496, 2013.
Article in English | MEDLINE | ID: mdl-23704989

ABSTRACT

The ability to efficiently generate integration-free induced pluripotent stem cells (iPSCs) from the most readily available source-peripheral blood-has the potential to expedite the advances of iPSC-based therapies. We have successfully generated integration-free iPSCs from cord blood (CB) CD34(+) cells with improved oriP/EBNA1-based episomal vectors (EV) using a strong spleen focus forming virus (SFFV) long terminal repeat (LTR) promoter. Here we show that Yamanaka factors (OCT4, SOX2, MYC, and KLF4)-expressing EV can also reprogram adult peripheral blood mononuclear cells (PBMNCs) into pluripotency, yet at a very low efficiency. We found that inclusion of BCL-XL increases the reprogramming efficiency by approximately 10-fold. Furthermore, culture of CD3(-)/CD19(-) cells or T/B cell-depleted MNCs for 4-6 days led to the generation of 20-30 iPSC colonies from 1 ml PB, an efficiency that is substantially higher than previously reported. PB iPSCs express pluripotency markers, form teratomas, and can be induced to differentiate in vitro into mesenchymal stem cells, cardiomyocytes, and hepatocytes. Used together, our optimized factor combination and reprogramming strategy lead to efficient generation of integration-free iPSCs from adult PB. This discovery has potential applications in iPSC banking, disease modeling and regenerative medicine.


Subject(s)
Blood Cells/cytology , Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/cytology , Transcription Factors/metabolism , bcl-X Protein/metabolism , Adult , Animals , Antigens, CD34/metabolism , Blood Cells/metabolism , Cell Differentiation , Cell Lineage , Cellular Reprogramming , Fetal Blood/cytology , Genetic Vectors/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Lentivirus/genetics , Mice , Plasmids/metabolism
6.
PLoS One ; 8(4): e59908, 2013.
Article in English | MEDLINE | ID: mdl-23573220

ABSTRACT

The effect of the cellular reprogramming process per se on mutation load remains unclear. To address this issue, we performed whole exome sequencing analysis of induced pluripotent stem cells (iPSCs) reprogrammed from human cord blood (CB) CD34(+) cells. Cells from a single donor and improved lentiviral vectors for high-efficiency (2-14%) reprogramming were used to examine the effects of three different combinations of reprogramming factors: OCT4 and SOX2 (OS), OS and ZSCAN4 (OSZ), OS and MYC and KLF4 (OSMK). Five clones from each group were subject to whole exome sequencing analysis. We identified 14, 11, and 9 single nucleotide variations (SNVs), in exomes, including untranslated regions (UTR), in the five clones of OSMK, OS, and OSZ iPSC lines. Only 8, 7, and 4 of these, respectively, were protein-coding mutations. An average of 1.3 coding mutations per CB iPSC line is remarkably lower than previous studies using fibroblasts and low-efficiency reprogramming approaches. These data demonstrate that point nucleotide mutations during cord blood reprogramming are negligible and that the inclusion of genome stabilizers like ZSCAN4 during reprogramming may further decrease reprogramming-associated mutations. Our findings provide evidence that CB is a superior source of cells for iPSC banking.


Subject(s)
Fetal Blood/cytology , Induced Pluripotent Stem Cells/physiology , Point Mutation , Animals , Cell Dedifferentiation , Cells, Cultured , Coculture Techniques , DNA Mutational Analysis , Exome , Genomic Instability , Humans , Induced Pluripotent Stem Cells/transplantation , Kruppel-Like Factor 4 , Mice , Mice, Inbred NOD , Mice, SCID , Open Reading Frames , Regenerative Medicine , Teratoma/pathology
7.
Cell Res ; 23(5): 658-72, 2013 May.
Article in English | MEDLINE | ID: mdl-23478301

ABSTRACT

The direct conversion of skin cells into somatic stem cells has opened new therapeutic possibilities in regenerative medicine. Here, we show that human induced mesenchymal stem cells (iMSCs) can be efficiently generated from cord blood (CB)- or adult peripheral blood (PB)-CD34(+) cells by direct reprogramming with a single factor, OCT4. In the presence of a GSK3 inhibitor, 16% of the OCT4-transduced CD34(+) cells are converted into iMSCs within 2 weeks. Efficient direct reprogramming is achieved with both episomal vector-mediated transient OCT4 expression and lentiviral vector-mediated OCT4 transduction. The iMSCs express MSC markers, resemble bone marrow (BM)-MSCs in morphology, and possess in vitro multilineage differentiation capacity, yet have a greater proliferative capacity compared with BM-MSCs. Similar to BM-MSCs, the implanted iMSCs form bone and connective tissues, and are non-tumorigenic in mice. However, BM-MSCs do not, whereas iMSCs do form muscle fibers, indicating a potential functional advantage of iMSCs. In addition, we observed that a high level of OCT4 expression is required for the initial reprogramming and the optimal iMSC self-renewal, while a reduction of OCT4 expression is required for multilineage differentiation. Our method will contribute to the generation of patient-specific iMSCs, which could have applications in regenerative medicine. This discovery may also facilitate the development of strategies for direct conversion of blood cells into other types of cells of clinical importance.


Subject(s)
Antigens, CD34/metabolism , Blood Cells/cytology , Fetal Blood/cytology , Mesenchymal Stem Cells/cytology , Octamer Transcription Factor-3/metabolism , Animals , Blood Cells/metabolism , Bone and Bones/pathology , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Connective Tissue/pathology , Fetal Blood/metabolism , Genetic Vectors/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Hematopoietic Stem Cells/cytology , Humans , Karyotyping , Lentivirus/genetics , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Octamer Transcription Factor-3/genetics , Transduction, Genetic
8.
Mol Ther ; 20(2): 408-16, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22108860

ABSTRACT

The reprogramming of cord blood (CB) cells into induced pluripotent stem cells (iPSCs) has potential applications in regenerative medicine by converting CB banks into iPSC banks for allogeneic cell replacement therapy. Therefore, further investigation into novel approaches for efficient reprogramming is necessary. Here, we show that the lentiviral expression of OCT4 together with SOX2 (OS) driven by a strong spleen focus-forming virus (SFFV) promoter in a single vector can convert 2% of CB CD34(+) cells into iPSCs without additional reprogramming factors. Reprogramming efficiency was found to be critically dependent upon expression levels of OS. To generate transgene-free iPSCs, we developed an improved episomal vector with a woodchuck post-transcriptional regulatory element (Wpre) that increases transgene expression by 50%. With this vector, we successfully generated transgene-free iPSCs using OS alone. In conclusion, high-level expression of OS alone is sufficient for efficient reprogramming of CB CD34(+) cells into iPSCs. This report is the first to describe the generation of transgene-free iPSCs with the use of OCT4 and SOX2 alone. These findings have important implications for the clinical applications of iPSCs.


Subject(s)
Cell Differentiation/genetics , Hematopoietic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Octamer Transcription Factor-3/genetics , SOXB1 Transcription Factors/genetics , Antigens, CD34/metabolism , Fetal Blood/cytology , Gene Expression , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Lentivirus/genetics , Proto-Oncogene Proteins c-myc/metabolism
9.
Mol Ther ; 19(10): 1896-904, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21772255

ABSTRACT

Current methods for eradicating clinically significant inhibitory antibodies to human factor VIII (hFVIII) in patients with hemophilia A rely on repeated delivery of high doses of factor concentrates for a minimum of many months. We hypothesize that tolerance can be induced more efficiently and reliably through hFVIII antigen presentation by tolerogenic dendritic cells (tDCs). In this study, we generated tDCs from hemophilia A mice and modified them with a foamy virus vector expressing a bioengineered hFVIII transgene. Naive and preimmunized mice infused with hFVIII expressing tDCs showed suppression of the T cell and inhibitor responses to recombinant hFVIII (rhFVIII). Treatment with hFVIII expressing tDCs was also associated with a higher percentage of splenocytes demonstrating a regulatory T cell phenotype in immunized mice. Furthermore, CD4(+) T cells harvested from recipients of hFVIII expression vector-modified tDCs were able to mediate antigen-specific immune suppression in naive secondary recipients. We also demonstrated a trend for improved suppression of inhibitor formation by coexpressing interleukin-10 (IL-10) and hFVIII from a bicistronic vector. These preclinical results demonstrate the potential for employing vector modified ex vivo generated tDCs to treat high titer inhibitors in patients with hemophilia A.


Subject(s)
Dendritic Cells/immunology , Factor VIII/antagonists & inhibitors , Hemophilia A/immunology , Transgenes , Animals , Factor VIII/immunology , Mice , T-Lymphocytes, Regulatory/immunology
10.
World J Biol Chem ; 2(6): 119-25, 2011 Jun 26.
Article in English | MEDLINE | ID: mdl-21765977

ABSTRACT

Ikaros (also known as Lyf-1) was initially described as a lymphoid-specific transcription factor. Although Ikaros has been shown to regulate hematopoietic stem cell renewal, as well as the development and function of cells from multiple hematopoietic lineages, including the myeloid lineage, Ikaros has primarily been studied in context of lymphoid development and malignancy. This review focuses on the role of Ikaros in myeloid cells. We address the importance of post-transcriptional regulation of Ikaros function; the emerging role of Ikaros in myeloid malignancy; Ikaros as a regulator of myeloid differentiation and function; and the selective expression of Ikaros isoform-x in cells with myeloid potential. We highlight the challenges of dissecting Ikaros function in lineage commitment decisions among lymphoid-myeloid progenitors that have emerged as a major myeloid differentiation pathway in recent studies, which leads to reconstruction of the traditional map of murine and human hematopoiesis.

11.
Mol Ther ; 18(1): 214-22, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19755963

ABSTRACT

The development of inhibitory antibodies to factor VIII (FVIII) is currently the most significant complication of FVIII replacement therapy in the management of patients with severe hemophilia A. Immune tolerance protocols for the eradication of inhibitors require daily delivery of intravenous FVIII for at least 6 months and are unsuccessful in 20-40% of treated patients. We hypothesize that tolerance can be induced more efficiently and reliably by delivery of FVIII antigen within autologous apoptotic cells (ACs). In this study, we demonstrated suppression of the T cell and inhibitor responses to FVIII by infusion of FVIII expression vector modified apoptotic syngeneic fibroblasts in both naive and preimmunized hemophilia A mice. ACs without FVIII antigen exerted modest generalized immune suppression mediated by anti-inflammatory signals. However, FVIII expressing apoptotic syngeneic fibroblasts produced much stronger antigen-specific immune suppression. Mice treated with these fibroblasts generated CD4+ T cells that suppressed the immune response to FVIII after adoptive transfer into naive recipients and antigen-specific CD4+CD25+ regulatory T cells (Tregs) that inhibited the proliferation of FVIII responsive effector T cells in vitro. These preclinical results demonstrate the potential for using FVIII vector modified autologous ACs to treat high-titer inhibitors in patients with hemophilia A.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Factor VIII/metabolism , Fibroblasts/metabolism , Fibroblasts/transplantation , Hemophilia A/therapy , Animals , Apoptosis/physiology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Factor VIII/genetics , Factor VIII/immunology , Fibroblasts/immunology , Fibroblasts/physiology , Interleukin-2 Receptor alpha Subunit/immunology , Mice , Mice, Knockout , Spleen/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
12.
Stem Cells Dev ; 14(2): 223-30, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15910249

ABSTRACT

Stem cell expansion remains an elusive but highly desirable goal. Here we show that platelet-derived growth factor (PDGF), along with cultured endothelial or stromal cells, significantly enhances expansion of human CD34+ cells in vitro. In media supplemented with thrombopoietin, stem cell factor, flt-3 ligand, and granulocyte-colony stimulating factor, CD34+ cells, as well as CFU-GM, BFU-CFU-E, CFU-GEMM, and CFU-MK, increased by 34.3-, 138-, 59.7-, 38.4-, and 86.0-fold, respectively. Co-culturing of CD34+ cells with cultured stromal cells or human umbilical cord vein endothelial cells (HUVECs) greatly enhanced expansion efficiency. The presence of PDGF (50 ng/ml) further augmented expansion, such that increases of 77.0-, 262-, 90.0-, 93.0-, and 200-fold, respectively, were achieved. Six weeks after infusion of expanded cells into NOD/SCID mice, human CD45+ cells were detected in recipients' bone marrow, spleen, and peripheral blood. Our results provide a rationale for development of a stem cell expansion protocol for clinical applications.


Subject(s)
Antigens, CD34/biosynthesis , Fetal Blood/cytology , Platelet-Derived Growth Factor/physiology , Umbilical Veins/metabolism , Animals , Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Cell Membrane/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques , Culture Media, Serum-Free/pharmacology , Endothelium, Vascular/cytology , Flow Cytometry , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Hematopoietic System , Humans , Integrin beta3/biosynthesis , Leukocyte Common Antigens/biosynthesis , Lipopolysaccharide Receptors/biosynthesis , Megakaryocytes/cytology , Mice , Mice, Inbred NOD , Mice, SCID , Platelet-Derived Growth Factor/metabolism , Receptors, CXCR4/metabolism , Spleen/cytology , Stem Cells/cytology , Stem Cells/metabolism , Stromal Cells/cytology , Time Factors , Umbilical Veins/cytology
13.
Br J Haematol ; 117(3): 735-46, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12028051

ABSTRACT

Platelet-derived growth factor (PDGF) is a major mitogen for connective tissue cells. In this study, we investigated the effects and mechanism of PDGF on the ex vivo expansion of cord blood CD34+ cells. Our data demonstrated that among various cytokine combinations of thrombopoietin (TPO), interleukin 1 beta (IL-1beta), IL-3, IL-6 and Flt-3 ligand (Flt-3L), TPO + IL-6 + Flt-3L was most efficient in promoting the expansion of CD34+ cells, CD34+CD38- cells, mixed-lineage colony-forming units (CFU-GEMM) and long-term culture-initiating cells (LTC-IC) by 21.7 +/- 5.00-, 103 +/- 27.9-, 10.7 +/- 7.94- and 6.52 +/- 1.51-fold, respectively, after 12-14 d of culture. The addition of PDGF increased the yield of these early progenitors by 45.0%, 66.5%, 45.1% and 79.8% respectively. More significantly, PDGF enhanced the engraftment of human CD45+ cells and their myeloid subsets (CD33+, CD14+ cells) in non-obese diabetic (NOD)/severe-combined immunodeficient (SCID) mice. The expression of PDGF receptor (PDGFR)-beta was not detectable in fresh CD34+ cells but was upregulated after culture for 3 d. PDGF also enhanced the development of adherent cells/clusters that expressed the endothelial markers VE-cadherin and CD31. These findings suggest that PDGF is an effective cytokine for the ex vivo expansion of early stem and progenitor cells. The mechanism could be mediated by PDGFR-beta on committed CD34+ progenitor cells and/or secondary to the stimulation of autologous, stromal feeder cells.


Subject(s)
Antigens, CD34/blood , Diabetes Mellitus, Experimental/therapy , Hematopoietic Stem Cells/drug effects , Platelet-Derived Growth Factor/pharmacology , Severe Combined Immunodeficiency/therapy , Animals , Cell Adhesion/drug effects , Cell Division/drug effects , Colony-Forming Units Assay , Cytokines/pharmacology , Fetal Blood/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Platelet-Derived Growth Factor/metabolism , Stem Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...