Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Nat Immunol ; 14(2): 162-71, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23263555

ABSTRACT

Signaling through the G protein-coupled receptors for the complement fragments C3a and C5a (C3aR and C5aR, respectively) by dendritic cells and CD4(+) cells provides costimulatory and survival signals to effector T cells. Here we found that when signals from C3aR and C5aR were not transduced into CD4(+) cells, signaling via the kinases PI(3)Kγ, Akt and mTOR ceased, activation of the kinase PKA increased, autoinductive signaling by transforming growth factor-ß1 (TGF-ß1) initiated and CD4(+) T cells became Foxp3(+) induced regulatory T cells (iT(reg) cells). Endogenous TGF-ß1 suppressed signaling through C3aR and C5aR by preventing the production of C3a and C5a and upregulating C5L2, an alternative receptor for C5a. The absence of signaling via C3aR and C5aR resulted in lower expression of costimulatory molecules and interleukin 6 (IL-6) and more production of IL-10. The resulting iT(reg) cells exerted robust suppression, had enhanced stability and suppressed ongoing autoimmune disease. Antagonism of C3aR and C5aR can also induce functional human iT(reg) cells.


Subject(s)
Forkhead Transcription Factors/metabolism , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Complement/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cell Communication/immunology , Cell Differentiation , Class Ib Phosphatidylinositol 3-Kinase/immunology , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Complement C3a/immunology , Complement C3a/metabolism , Complement C5a/immunology , Complement C5a/metabolism , Cyclic AMP-Dependent Protein Kinases/immunology , Cyclic AMP-Dependent Protein Kinases/metabolism , Forkhead Transcription Factors/immunology , Gene Expression Regulation , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptor Cross-Talk/immunology , Receptor, Anaphylatoxin C5a/immunology , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Receptors, Complement/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta1/immunology
2.
Am J Pathol ; 192(2): 361-378, 2022 02.
Article in English | MEDLINE | ID: mdl-35144762

ABSTRACT

As per the classical view of the coagulation system, it functions solely in plasma to maintain hemostasis. An experimental approach modeling vascular reconstitution was used to show that vascular endothelial cells (ECs) endogenously synthesize coagulation factors during angiogenesis. Intracellular thrombin generated from this synthesis promotes the mitotic function of vascular endothelial cell growth factor A (VEGF-A). The thrombin concurrently cleaves C5a from EC-synthesized complement component C5 and unmasks the tethered ligand for EC-expressed protease-activated receptor 4 (PAR4). The two ligands jointly trigger EC C5a receptor-1 (C5ar1) and PAR4 signaling, which together promote VEGF receptor 2 growth signaling. C5ar1 is functionally associated with PAR4, enabling C5a or thrombin to elicit Gαi and/or Gαq signaling. EC coagulation factor and EC complement component synthesis concurrently down-regulate with contact inhibition. The connection of these processes with VEGF receptor 2 signaling provides new insights into mechanisms underlying angiogenesis. Knowledge of endogenous coagulation factor/complement component synthesis and joint PAR4/C5ar1 signaling could be applied to other cell types.


Subject(s)
Blood Coagulation Factors/biosynthesis , Endothelial Cells/metabolism , Neovascularization, Physiologic , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Proteinase-Activated/metabolism , Signal Transduction , Animals , Blood Coagulation Factors/genetics , Female , Male , Mice , Mice, Knockout , Receptor, Anaphylatoxin C5a/genetics , Receptors, Proteinase-Activated/genetics
3.
Am J Pathol ; 189(7): 1386-1401, 2019 07.
Article in English | MEDLINE | ID: mdl-31103439

ABSTRACT

Recent studies traced inflammatory bowel disease in some patients to deficiency of CD55 [decay-accelerating factor (DAF)], but the mechanism underlying the linkage remained unclear. Herein, we studied the importance of DAF in enabling processes that program tolerance in the gut and the eye, two immune-privileged sites where immunosuppressive responses are continuously elicited. Unlike oral feeding or ocular injection of ovalbumin in wild-type (WT) mice, which induced dominant immune tolerance, identical treatment of DAF-/- mice or DAF-/- to WT bone marrow chimeras did not. While 10% to 30% of mesenteric and submandibular lymph node CD4+ cells became robust T-regulatory cells (Tregs) in WT forkhead box P3 (Foxp3)-green fluorescent protein mice, few in either site became Tregs with little suppressor activity in DAF-/- Foxp3-green fluorescent protein mice. Phenotyping of CD103+ dendritic cells (DCs) from the ovalbumin-fed DAF-/- mice showed impaired expression of inducer of costimulation (ICOS) ligand, programmed death receptor 1-ligand 1 (PD1-L1), CxxxC chemokine receptor 1 (Cx3CR1), CCR7, and CCR9. Analyses of elicited DAF-/- Foxp3+ Tregs showed reduced expression of interferon regulatory factor 8 (IRF-8)/aldehyde dehydrogenase 1 family member A2 (Aldh1a2) and glycoprotein A repetitions predominant/latency-associated protein associated with Treg transforming growth factor-ß production and presentation, as well as integrin ß6/integrin ß8 associated with Treg and CD103+ DC transforming growth factor-ß release. Thus, DAF is required for the properties of CD103+ DCs and their naïve CD4+ cell partners that together program tolerance.


Subject(s)
Antigens, CD/immunology , Autoimmune Diseases/immunology , CD55 Antigens/immunology , Dendritic Cells/immunology , Immune Tolerance , Integrin alpha Chains/immunology , Animals , Antigens, CD/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Autoimmune Diseases/prevention & control , CD55 Antigens/genetics , Dendritic Cells/pathology , Integrin alpha Chains/genetics , Integrin beta Chains/genetics , Integrin beta Chains/immunology , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
4.
Immunity ; 28(3): 425-35, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18328742

ABSTRACT

Costimulatory signals are critical to T cell activation, but how their effects are mediated remains incompletely characterized. Here, we demonstrate that locally produced C5a and C3a anaphylatoxins interacting with their G protein-coupled receptors (GPCRs), C5aR and C3aR, on APCs and T cells both upstream and downstream of CD28 and CD40L signaling are integrally involved in T cell proliferation and differentiation. Disabling these interactions reduced MHC class II and costimulatory-molecule expression and dramatically diminished T cell responses. Importantly, impaired T cell activation by Cd80-/-Cd86-/- and Cd40-/- APCs was reconstituted by added C5a or C3a. C5aR and C3aR mediated their effects via PI-3 kinase-gamma-dependent AKT phosphorylation, providing a link between GPCR signaling, CD28 costimulation, and T cell survival. These local paracrine and autocrine interactions thus operate constitutively in naive T cells to maintain viability, and their amplification by cognate APC partners thus is critical to T cell costimulation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Survival/immunology , Complement C3a/immunology , Complement C5a/immunology , Lymphocyte Activation/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Ligand/immunology , CD40 Ligand/metabolism , Cell Differentiation/immunology , Complement C3a/metabolism , Complement C5a/metabolism , Flow Cytometry , Immunoblotting , Immunoprecipitation , Mice , Mice, Transgenic , Polymerase Chain Reaction , RNA, Messenger/analysis , Receptor, Anaphylatoxin C5a/immunology , Receptor, Anaphylatoxin C5a/metabolism , Signal Transduction/immunology
5.
Front Immunol ; 14: 1290684, 2023.
Article in English | MEDLINE | ID: mdl-38406578

ABSTRACT

The transcription factor Kruppel-like factor 4 (KLF4) regulates the expression of immunosuppressive and anti-thrombotic proteins. Despite its importance in maintaining homeostasis, the signals that control its expression and the mechanism of its transactivation remain unclarified. CD55 [aka decay accelerating factor (DAF)], now known to be a regulator of T and B cell responses, biases between pro- and anti-inflammatory processes by controlling autocrine C3a and C5a receptor (C3ar1/C5ar1) signaling in cells. The similarity in CD55's and KLF4's regulatory effects prompted analyses of their functional relationship. In vascular endothelial cells (ECs), CD55 upregulation accompanied KLF4 expression via a p-CREB and CREB Binding Protein (CBP) mechanism. In both ECs and macrophages, CD55 expression was essential for KLF4's downregulation of pro-inflammatory/pro-coagulant proteins and upregulation of homeostatic proteins. Mechanistic studies showed that upregulation of KLF4 upregulated CD55. The upregulated CD55 in turn enabled the recruitment of p-CREB and CBP to KLF4 needed for its transcription. Activation of adenylyl cyclase resulting from repression of autocrine C3ar1/C5ar1 signaling by upregulated CD55 concurrently led to p-CREB and CBP recruitment to KLF4-regulated genes, thereby conferring KLF4's transactivation. Accordingly, silencing CD55 in statin-treated HUVEC disabled CBP transfer from the E-selectin to the eNOS promoter. Importantly, silencing CD55 downregulated KLF4's expression. It did the same in untreated HUVEC transitioning from KLF4low growth to KLF4hi contact inhibition. KLF4's and CD55's function in ECs and macrophages thus are linked via a novel mechanism of gene transactivation. Because the two proteins are co-expressed in many cell types, CD55's activity may be broadly tied to KLF4's immunosuppressive and antithrombotic activities.


Subject(s)
Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Endothelial Cells/metabolism , Up-Regulation , Promoter Regions, Genetic
6.
PLoS One ; 6(10): e24431, 2011.
Article in English | MEDLINE | ID: mdl-21984892

ABSTRACT

BACKGROUND: In addition to its complement-regulating activity, CD55 is a ligand of the adhesion class G protein-coupled receptor CD97; however, the relevance of this interaction has remained elusive. We previously showed that mice lacking a functional CD97 gene have increased numbers of granulocytes. METHODOLOGY/RESULTS: Here, we demonstrate that CD55-deficient mice display a comparable phenotype with about two-fold more circulating granulocytes in the blood stream, the marginated pool, and the spleen. This granulocytosis was independent of increased complement activity. Augmented numbers of Gr-1-positive cells in cell cycle in the bone marrow indicated a higher granulopoietic activity in mice lacking either CD55 or CD97. Concomitant with the increase in blood granulocyte numbers, Cd55⁻/⁻ mice challenged with the respiratory pathogen Streptococcus pneumoniae developed less bacteremia and died later after infection. CONCLUSIONS: Collectively, these data suggest that complement-independent interaction of CD55 with CD97 is functionally relevant and involved in granulocyte homeostasis and host defense.


Subject(s)
CD55 Antigens/metabolism , Granulocytes/immunology , Homeostasis/immunology , Host-Pathogen Interactions/immunology , Streptococcus pneumoniae/immunology , Animals , Cell Movement/immunology , Complement System Proteins/immunology , Disease Resistance/immunology , Granulocytes/cytology , Leukocyte Count , Membrane Glycoproteins/metabolism , Mice , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/microbiology , Receptors, G-Protein-Coupled
7.
Invest Ophthalmol Vis Sci ; 50(8): 3778-82, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19443714

ABSTRACT

PURPOSE: To investigate the role of decay-accelerating factor (DAF), a cell surface complement regulator that recently has been linked to T-cell responses and autoimmunity in the pathogenesis of experimental autoimmune uveitis (EAU). METHODS: EAU was induced in wild-type (WT) and Daf1(-/-) mice, and their disease severities, IRBP specific Th1/Th17 responses, and cytokine expression profiles were compared. In a test of the efficacy of treatment with soluble mouse DAF protein, EAU was induced in disease-susceptible B10.RIII mice, and they were treated with 0.5 mg soluble DAF protein or equal volume of PBS IP every other day. Retinal histology and IRBP-specific T-cell responses were compared after 14 days. RESULTS: Both EAU incidence and histopathology scores were significantly greater in Daf1(-/-) mice. There was a >10-fold greater mononuclear cell influx into the retina together with severe vasculitic lesions, retinal folding, and photoreceptor cell layer destruction. There were 5- to 7-fold greater Th1 and 3- to 4-fold greater Th17 responses against IRBP in Daf1(-/-) mice with EAU, and they expressed significantly elevated levels of GM-CSF, IL-2, IL-3, and IFN-gamma. WT B10.RIII mice that received soluble DAF protein treatments exhibited decreased IRBP-specific Th1/Th17 responses and were protected from retinal injury compared with the mice that received PBS treatments. CONCLUSIONS: DAF significantly influences IRBP-specific Th1 and Th17 responses and disease severity in EAU. Systemic upregulation of DAF levels could be used to suppress retinal antigen(s)-specific autoimmunity to treat autoimmune posterior uveitis.


Subject(s)
Autoimmune Diseases/prevention & control , Autoimmunity/physiology , CD55 Antigens/physiology , Th1 Cells/immunology , Uveitis, Posterior/prevention & control , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Disease Models, Animal , Eye Proteins , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins , Retinol-Binding Proteins , Uveitis, Posterior/immunology , Uveitis, Posterior/pathology
8.
J Immunol ; 180(9): 5882-9, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18424707

ABSTRACT

IFN-gamma- and IL-17-producing T cells autoreactive across myelin components are central to the pathogenesis of multiple sclerosis. Using direct in vivo, adoptive transfer, and in vitro systems, we show in this study that the generation of these effectors in myelin oligodendrocyte glycoprotein(35-55)-induced experimental autoimmune encephalomyelitis depends on interactions of locally produced C3a/C5a with APC and T cell C3aR/C5aR. In the absence of the cell surface C3/C5 convertase inhibitor decay-accelerating factor (DAF), but not the combined absence of DAF and C5aR and/or C3aR on APC and T cells, a heightened local autoimmune response occurs in which myelin destruction is markedly augmented in concert with markedly more IFN-gamma(+) and IL-17(+) T cell generation. The augmented T cell response is due to increased IL-12 and IL-23 elaboration by APCs together with increased T cell expression of the receptors for each cytokine. The results apply to initial generation of the IL-17 phenotype because naive CD62L(high) Daf1(-/-) T cells produce 3-fold more IL-17 in response to TGF-beta and IL-6, whereas CD62L(high) Daf1(-/-)C5aR(-/-)C3aR(-/-) T cells produce 4-fold less.


Subject(s)
Antigen-Presenting Cells/immunology , Complement C3a/immunology , Complement C5a/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interferon-gamma/immunology , Interleukin-17/immunology , Multiple Sclerosis/immunology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/metabolism , CD55 Antigens/immunology , CD55 Antigens/metabolism , Complement C3-C5 Convertases/immunology , Complement C3-C5 Convertases/metabolism , Complement C3a/metabolism , Complement C5a/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Glycoproteins/immunology , Glycoproteins/pharmacology , Interferon-gamma/biosynthesis , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-17/biosynthesis , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Mice , Mice, Knockout , Multiple Sclerosis/metabolism , Myelin Sheath/immunology , Myelin Sheath/metabolism , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments/immunology , Peptide Fragments/pharmacology , Rats , Receptor, Anaphylatoxin C5a/immunology , Receptor, Anaphylatoxin C5a/metabolism , T-Lymphocytes/metabolism
9.
J Virol ; 80(10): 4833-46, 2006 May.
Article in English | MEDLINE | ID: mdl-16641275

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman's disease. Most KS tumor cells are latently infected with KSHV and are of endothelial origin. While PEL-derived cell lines maintain KSHV indefinitely, all KS tumor-derived cells to date have lost viral genomes upon ex vivo cultivation. To study KSHV latency and tumorigenesis in endothelial cells, we generated telomerase-immortalized human umbilical vein endothelial (TIVE) cells. TIVE cells express all KSHV latent genes 48 h postinfection, and productive lytic replication could be induced by RTA/Orf50. Similar to prior models, infected cultures gradually lost viral episomes. However, we also obtained, for the first time, two endothelial cell lines in which KSHV episomes were maintained indefinitely in the absence of selection. Long-term KSHV maintenance correlated with loss of reactivation in response to RTA/Orf50 and complete oncogenic transformation. Long-term-infected TIVE cells (LTC) grew in soft agar and proliferated under reduced-serum conditions. LTC, but not parental TIVE cells, formed tumors in nude mice. These tumors expressed high levels of the latency-associated nuclear antigen (LANA) and expressed lymphatic endothelial specific antigens as found in KS (LYVE-1). Furthermore, host genes, like those encoding interleukin 6, vascular endothelial growth factor, and basic fibroblast growth factor, known to be highly expressed in KS lesions were also induced in LTC-derived tumors. KSHV-infected LTCs represent the first xenograft model for KS and should be of use to study KS pathogenesis and for the validation of anti-KS drug candidates.


Subject(s)
Endothelial Cells/virology , Endothelium, Vascular/virology , Herpesviridae/physiology , Sarcoma, Kaposi/virology , Telomerase/physiology , Virus Latency/physiology , Animals , Cell Culture Techniques/methods , Cell Line, Transformed , Cytokines/biosynthesis , Cytokines/genetics , Disease Susceptibility , Endothelial Cells/cytology , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Gene Expression Regulation, Viral , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Virus Replication/physiology
10.
J Biol Chem ; 280(5): 3862-74, 2005 Feb 04.
Article in English | MEDLINE | ID: mdl-15525642

ABSTRACT

Latently infected Kaposi's sarcoma-associated herpes-virus (KSHV)-associated tumor cells have both endothelial and lymphoid origins and express a limited set of latent viral genes. One such gene, ORF73, encodes the latency-associated nuclear antigen (LANA), a multifunctional protein that plays roles in viral DNA replication, episome maintenance, and transcriptional regulation. LANA interacts with cellular proteins involved in transcriptional regulation such as the tumor suppressors, retinoblastoma (Rb) and p53, and RING3 family members. Although several reports about specific LANA-regulated promoters exist, only limited data are available that address how LANA expression in KSHV-infected cells globally affects cellular gene expression, thereby potentially contributing to KSHV pathogenicity. To investigate this question, we generated an Epstein-Barr virus-negative Burkitts lymphoma line that expresses LANA from a tetracycline-inducible promoter (BJAB/Tet-On/LANA), and we performed microarray-based gene expression profiling. Expression profiling at different time points post-induction revealed that 186 genes were activated or repressed over 2-fold in the presence of LANA. Of these genes, 41 are regulated in the Rb/E2F pathway, whereas 7 are related to p53 signaling. To determine whether these gene expression changes translate into LANA-dependent changes in cell cycle regulation, we overexpressed p16 INK4a, a CDK4/6 inhibitor that efficiently induces cell cycle arrest in Rb-positive cells. Under these conditions, LANA expression protects lymphoid cells from p16 INK4a-induced cell cycle arrest and induces S-phase entry.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Expression Profiling , Herpesvirus 8, Human/genetics , Lymphocytes/virology , Nuclear Proteins/genetics , Antigens, Viral , Burkitt Lymphoma , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , E2F Transcription Factors , Gene Expression Regulation, Viral , Humans , Lymphocytes/cytology , Retinoblastoma Protein/metabolism , S Phase/physiology , Trans-Activators/metabolism , Transcription Factors/metabolism , Transfection , beta Catenin
11.
Oncology ; 63(3): 286-96, 2002.
Article in English | MEDLINE | ID: mdl-12381909

ABSTRACT

Liver metastasis is one of the poor prognostic factors for gastric cancer. Hepatocyte growth factor (HGF) and its receptor, c-Met, have been reported to be related to the proliferation of carcinoma cells. We examined c-Met and HGF expression in stage IV gastric cancers (n = 121) and compared the results in groups with liver metastasis (n = 47, LM group) and without liver metastasis (n = 74, no-LM group). The survival rate for the LM group was significantly poorer than for the no-LM group (p < 0.01). We found a high frequency of c-Met expression in the LM group compared with the no-LM group at protein level detected by immunohistochemistry (p = 0.0005) and at mRNA level detected by semiquantitative reverse transcriptase-polymerase chain reaction (p = 0.0386) in primary gastric tumors. Furthermore, we evaluated HGF expression in both carcinoma cells and stromal cells in gastric cancers. There was no significant difference in the HGF expression between the LM and no-LM groups. The labeling index of proliferating cell nuclear antigen for the carcinomas in the LM group was higher than that in the no-LM group (47.1 +/- 24.5 vs. 26.2 +/- 24.5%, p < 0.0001). Thus, the high frequency of c-Met overexpression in carcinoma cells may be involved in the mechanism of liver metastasis in gastric cancers. Moreover, the evaluation of c-Met expression might be a useful indicator of liver metastasis in patients with gastric cancer.


Subject(s)
Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Proto-Oncogene Proteins c-met/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/secondary , Adult , Aged , Aged, 80 and over , Carcinoma, Signet Ring Cell/genetics , Carcinoma, Signet Ring Cell/metabolism , Carcinoma, Signet Ring Cell/secondary , DNA Primers/chemistry , Female , Hepatocyte Growth Factor/metabolism , Humans , Immunoenzyme Techniques , Liver Neoplasms/genetics , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-met/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/genetics , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL