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











Publication year range
1.
Hum Immunol ; 84(8): 359-365, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37169599

ABSTRACT

Human amnion epithelial cells (hAEC) can be efficiently isolated from full-term amnion membrane and have been gaining recognition as advanced medical products. Such cells originate directly from the embryo during the early phase of development and exert a crucial function in the establishment of a tolerogenic environment, to avoid maternal immune rejection. Amnion cell immuno-modulation may be exploited, but additional efforts are required to establish the mechanisms underlying such capacity. The way to fully clarify such an issue is so far long. Here we overview current knowledge on the effects on innate or adaptive immune cells offered by intact hAEC or secreted mediators, pinpointing the mechanisms to date elucidated by our group and others. We move from the description of hAEC general features to molecular intermediaries generating effects directly or indirectly on immune cells. We focus on the role of non-canonical HLA class I molecules, with emphasis on HLA-G, but expand such analysis on adenosinergic mediators, cytokines, and hAEC-derived microvesicles. Finally, we report the ongoing clinical trials exploiting hAEC multipotency and immune modulation.


Subject(s)
Epithelial Cells , HLA-G Antigens , Humans , Amnion , Cells, Cultured , Cytokines
2.
Immunol Lett ; 205: 40-50, 2019 01.
Article in English | MEDLINE | ID: mdl-30447309

ABSTRACT

Human myeloma cells grow in a hypoxic acidic niche in the bone marrow. Cross talk among cellular components of this closed niche generates extracellular adenosine, which promotes tumor cell survival. This is achieved through the binding of adenosine to purinergic receptors into complexes that function as an autocrine/paracrine signal factor with immune regulatory activities that i) down-regulate the functions of most immune effector cells and ii) enhance the activity of cells that suppress anti-tumor immune responses, thus facilitating the escape of malignant myeloma cells from immune surveillance. Here we review recent findings confirming that the dominant phenotype for survival of tumor cells is that where the malignant cells have been metabolically reprogrammed for the generation of lactic acidosis in the bone marrow niche. Adenosine triphosphate and nicotinamide-adenine dinucleotide extruded from tumor cells, along with cyclic adenosine monophosphate, are the main intracellular energetic/messenger molecules that serve as leading substrates in the extracellular space for membrane-bound ectonucleotidases metabolizing purine nucleotides to signaling adenosine. Within this mechanistic framework, the adenosinergic substrate conversion can vary significantly according to the metabolic environment. Indeed, the neoplastic expansion of plasma cells exploits both enzymatic networks and hypoxic acidic conditions for migrating and homing to a protected niche and for evading the immune response. The expression of multiple specific adenosine receptors in the niche completes the profile of a complex regulatory framework whose signals modify multiple myeloma and host immune responses.


Subject(s)
Adenine Nucleotides/metabolism , Adenosine/metabolism , Antigens, CD/metabolism , Bone Marrow Cells/enzymology , Multiple Myeloma/enzymology , Multiple Myeloma/pathology , Bone Marrow Cells/immunology , Humans , Immunosuppression Therapy , Multiple Myeloma/immunology , Receptors, Purinergic/metabolism , Signal Transduction/immunology , Tumor Microenvironment/immunology
3.
Immunol Lett ; 205: 25-30, 2019 01.
Article in English | MEDLINE | ID: mdl-29550257

ABSTRACT

Adenosine (ADO) is an immunosuppressive molecule with multiple functions in different human organs. ADO is released through the concerted action of surface molecules endowed with enzymatic functions, that belong to two different adenosinergic pathways. The canonical pathway is started by CD39, that converts ATP to AMP. On the other hand, the non-canonical pathway metabolizes NAD+ to ADPR, through the action of CD38. The latter byproduct is then converted to AMP by CD203a/PC-1. Both pathways converge to CD73, that fully degrades AMP to the final product ADO. In this Review we take into account the most relevant finding regarding the expression of ectoenzymes belonging to both adenosinergic pathways in different cell types, including regulatory cell subsets and neoplastic cells. Moreover, we summarize the role of these molecules in different physiological and pathological settings. Finally, we discuss potential therapeutic application of specific inhibitors of ectoenzymes and/or ADO receptors.


Subject(s)
Adenosine/metabolism , Antigens, CD/metabolism , Receptors, Purinergic P1/metabolism , Adenosine Triphosphate/metabolism , Animals , Antigens, CD/genetics , Humans , Leukocytes/metabolism , NAD/metabolism , Neoplasms/metabolism , Receptors, Purinergic P1/genetics , Signal Transduction/immunology
4.
Oncoimmunology ; 7(8): e1458809, 2018.
Article in English | MEDLINE | ID: mdl-30221054

ABSTRACT

Multiple myeloma (MM) derives from malignant transformation of plasma cells (PC), which accumulate in the bone marrow (BM), where microenvironment supports tumor growth and inhibits anti-tumor immune responses. Adenosine (ADO), an immunosuppressive molecule, is produced within MM patients' BM by adenosinergic ectoenzymes, starting from ATP (CD39/CD73) or NAD+ [CD38/CD203a(PC-1)/CD73]. These ectoenzymes form a discontinuous network expressed by different BM cells. We investigated the expression and function of ectoenzymes on microvesicles (MVs) isolated from BM plasma samples of patients with MM, using asymptomatic forms of monoclonal gammopathy of undetermined significance (MGUS) and smoldering MM (SMM) as controls. The percentage of MVs expressing ectoenzymes at high levels was higher when derived from MM patients than controls. BM CD138+ PC from MM patients expressed high levels of all ectoenzymes. Paired MVs samples confirmed a higher percentage of MVs with high ectoenzymes expression in MM patients than controls. Pooled MVs from MM patients or controls were tested for ADO production. The catabolism of ATP, NAD+, ADPR and AMP to ADO was higher in MVs from MM patients than in those from controls. In conclusion, our results confirmed the hypothesis that MVs in MM niche are main contributor of ADO production. The ability of MVs to reach biological fluids strongly support the view that MVs may assume diagnostic and pathogenetic roles.

5.
Mediators Inflamm ; 2018: 7019398, 2018.
Article in English | MEDLINE | ID: mdl-29769837

ABSTRACT

Adenosine (ADO) is an immunosuppressive molecule, which suppresses the immune responses by interacting with specific receptors expressed by immune effector cells. ADO is produced from ATP through the enzymatic activities of CD39 and CD73. Alternatively, ADO can be generated starting from NAD+, which is metabolized by the concerted action of CD38, CD203a/PC-1, and CD73. The role of ADO in immunity has been characterized in the last years in physiology and in pathological settings. This review examines a panel of reports focused on the functions of ADO in the context of human autoimmune/inflammatory diseases and the selected animal models. The final aim is to consider the role of adenosinergic ectoenzymes and ADO receptors as novel therapeutic targets for selected diseases.


Subject(s)
Adenosine/metabolism , Autoimmune Diseases/metabolism , 5'-Nucleotidase/metabolism , ADP-ribosyl Cyclase 1/metabolism , Animals , Antigens, CD/metabolism , Apyrase/metabolism , Humans
6.
Cancer Gene Ther ; 18(5): 346-57, 2011 May.
Article in English | MEDLINE | ID: mdl-21233855

ABSTRACT

Cancer treatment is often complicated by resistance to conventional anti-cancer treatment and to more recently developed immunotherapy and gene therapy. These therapeutic modalities aim at activating death pathways within cancer cells. Attempts to activate the apoptotic death pathway, by overexpressing proapoptotic signals, are compromised by cancer defense mechanisms, which disrupt the apoptotic-signaling cascade downstream of the overexpressed component. Here, we describe a therapeutic option of triggering apoptosis without activating the apoptotic-signaling cascade or using the native apoptosis executioner nuclease. We have engineered Deoxyribonuclease-1 (DNase1), a waste-management enzyme, by deleting its signal peptide, adding a nuclear localization signal, and mutating its actin-binding site. Apoptosis studies and colony-forming assay for assessing cell viability were conducted in apoptosis-resistant Mel-Juso human melanoma cells. The modified DNase1 reduced cell viability by 77% relative to controls. It also induced typical microscopic features of cellular apoptosis, such as Terminal Transferase dUTP Nick-End Labeling-positive cells and DNA fragmentation. Quantification of apoptosis by Laser scanning cytometry demonstrated high-killing efficiency of 70-100%. The results suggest that this modified DNase1 can efficiently eliminate apoptosis-resistant cancer cells through apoptosis. Coupled to different tissue-specific gene expression elements, this recombinant DNase1 may serve as a platform for eliminating a variety of cancer types.


Subject(s)
Apoptosis/physiology , Deoxyribonuclease I/metabolism , Genetic Therapy/methods , Melanoma/therapy , Protein Engineering/methods , Signal Transduction/physiology , Apoptosis/genetics , Cell Line, Tumor , Colony-Forming Units Assay , DNA Fragmentation , Humans , In Situ Nick-End Labeling , Laser Scanning Cytometry , Melanoma/genetics , Signal Transduction/genetics
7.
Leukemia ; 24(5): 958-69, 2010 May.
Article in English | MEDLINE | ID: mdl-20220774

ABSTRACT

Homing of chronic lymphocytic leukemia (CLL) cells to sites favoring growth, a critical step in disease progression, is principally coordinated by the CXCL12/CXCR4 axis. A cohort of 62 CLL patients was divided into migrating and nonmigrating subsets according to chemotaxis toward CXCL12. Migrating patients phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2) proteins more than nonmigrating patients (P<0.0002). CD38 expression was the parameter most strongly associated with heightened CXCL12 signaling (P<0.0001), confirmed by independent statistical approaches. Consistent with this observation, CD38(-) CLL cells in samples with bimodal CD38 expression responded less to CXCL12 than the intact clone (P=0.003). Furthermore, lentivirus-induced de novo expression of CD38 was paralleled by increased responses to CXCL12, as compared with cells infected with a control virus. CD38 ligation with agonistic monoclonal antibodies (mAbs) enhanced CXCL12 signaling, whereas blocking anti-CD38 mAbs inhibited chemokine effects in vitro. This is attributed to physical proximity on the membrane between CD38 and CXCR4 (the CXCL12 receptor), as shown by (i) coimmunoprecipitation and (ii) confocal microscopy experiments. Blocking anti-CD38 mAbs significantly compromised homing of CLL cells from blood to lymphoid organs in a mouse model. These results indicate that CD38 synergizes with the CXCR4 pathway and support the working hypothesis that migration is a central step in disease progression.


Subject(s)
ADP-ribosyl Cyclase 1/physiology , Cell Movement , Chemokine CXCL12/metabolism , Chemotaxis , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Animals , Female , Humans , Immunoprecipitation , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Signal Transduction
9.
J Biol Regul Homeost Agents ; 19(3-4): 145-52, 2005.
Article in English | MEDLINE | ID: mdl-16602630

ABSTRACT

An anti-CD38 mAb (IB4) coupled to saporin-S6, a type 1 ribosome-inactivating protein (RIP), was designed for ex vivo or loco-regional therapeutical applications in myeloma and lymphoma. The ability of this immunotoxin to eliminate CD38+ cells was studied in vitro on selected CD38+ human cell lines (Raji, HBL6, L540 and CEM) and on CD38+ neoplastic cells from a Non Hodgkin Lymphoma (NHL) patient. HBL6, Raji and L540 cells resulted very sensitive to the IB4/saporin-S6 conjugate, concentrations as low as 100 pM of the immunotoxin completely inhibited protein synthesis. CD38+ neoplastic cells from the NHL patient were completely eliminated after treatment with immunotoxin at 10 nM concentration. CFU-c rescue by bone marrow precursors was maintained after exposure to the immunotoxin. These results indicate that IB4/saporin-S6 is endowed with strong and specific cytotoxic effects on selected CD38+ tumor cells lineages. Consequently, it is reasonable to propose a clinical use of the IB4/saporin-S6 for ex vivo purging of unwanted cells (e.g. depletion of contaminating neoplastic cells in aphereses obtained from G-CSF-treated patients) or for loco-regional therapies of CD38+ tumors.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Immunotoxins/immunology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Separation , Drug Design , Humans , Immunotoxins/pharmacology , In Vitro Techniques , N-Glycosyl Hydrolases/pharmacology , Plant Proteins/pharmacology , Protein Synthesis Inhibitors/pharmacology , Ribosome Inactivating Proteins, Type 1 , Saporins
11.
Biotechnol Adv ; 18(5): 385-401, 2000 Aug.
Article in English | MEDLINE | ID: mdl-14538101

ABSTRACT

This survey is an overview of the applications of murine, humanized and recombinant monoclonal antibodies for in vivo diagnostic and therapeutic applications. Monoclonal antibodies (mAb) have been applied to the diagnosis and therapy of an array of human diseases. The initial failures of early clinical trials have been overcome through the production of a new generation of mAb which features reduced immunogenicity and improved targeting abilities. The early models of mAb therapy were focused on enhancing the cytolytic mechanisms against the tumor cells. More recently, successful mAb-based therapies were targeted to molecules involved in the regulation of growth of cancer cells. This has highlighted the relevance of understanding receptor-mediated signaling events, and may provide new opportunities for anti-tumor antibody targeting. Despite all the difficulties, clinical data is outlining an increasingly significant role for antibody-mediated cancer therapy as a versatile and powerful instrument in cancer treatment. One reasonable expectation is that treatment at an earlier stage in the disease process or in minimal residual disease may be more advantageous.

12.
Exp Cell Res ; 247(2): 441-50, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10066372

ABSTRACT

Human monoclonal antibodies (HuMAb) specific for a 14-kDa perchloric acid-soluble protein (defined as UK114) were produced by somatic fusion of the human-mouse myeloma K6H6/B5 with Epstein-Barr virus-transformed peripheral B lymphocytes from a cancer patient previously treated with UK101 preparations, containing the UK114 protein. Three IgM-secreting clones were selected on the criteria of specificity for the purified UK114 protein immobilized onto plastic and adapted to grow in a serum-free medium. The reactivity of these antibodies showed a broad distribution pattern restricted to fresh tumor tissues and tumor cell lines, mainly of the adenocarcinoma type. None of the normal cells, nonmalignant cell lines, and normal tissues surrounding the neoplastic lesions were reactive. The immunochemical analysis of the target antigens showed that the HuMAb recognize a molecule of 220 kDa selectively expressed by the surface of tumor cells, as well as a cytoplasmic 14-kDa protein. The 220-kDa antigen was different from other tumor-associated antigens with similar molecular mass and, so far, unique. In the presence of human complement, two of three HuMAb are cytotoxic for tumor cells expressing the 220-kDa surface antigen. The tumor specificity and the lytic ability attributed to these HuMAb are promising features for the exploration of future clinical applications.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Antigens, Surface/immunology , Neoplasm Proteins/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Complement C8/immunology , Complement C9/immunology , HL-60 Cells , HT29 Cells , Humans , Immunoglobulins/immunology , Mice , Precipitin Tests , Tumor Cells, Cultured
13.
Biochem J ; 330 ( Pt 3): 1129-35, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9494077

ABSTRACT

Soluble forms of membrane receptors are emerging candidates as physiological regulators of leukocyte trafficking. In the present study, we found that the soluble form of the CD38 antigen (sCD38) bears a binding domain of low affinity for a cellular receptor on U937 cells. Cross-linking and peptide-mapping studies confirmed the physical association and the identification of the U937 receptor as a 130 kDa protein. The binding of sCD38 to the receptor was differentially inhibited by several monoclonal antibodies against the CD31 cell-adhesion molecule. Thus the interaction was analysed through direct association of soluble and membrane CD38 with soluble recombinant murine CD31 with three N-terminal and with all six extracellular Ig domains. Cross-linking experiments on U937 intact cells, and ligand blot assays of the immunoprecipitated CD38 molecule, indicated that (i) the recognized epitope is determined by the tertiary structure of the molecule, and that (ii) the binding domain involved resides in the ectocellular portion of the CD31 molecule, more precisely in the first three N-terminal domains. A comparative functional activity between murine and human CD31 was also explored. The data presented suggest that (i) human CD31 bears a highly functional similarity with its murine counterpart, as it is a receptor in myeloid cells with more than one ligand (the alphavbeta3 integrin and the CD38 molecule), and that (ii) the activity of sCD38 as decoy molecule for CD31 may play an important role in cell-cell interactions in physiological and pathological conditions.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation/metabolism , NAD+ Nucleosidase/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Animals , Binding Sites , Cross-Linking Reagents , Humans , Jurkat Cells , Kinetics , Leukocytes/physiology , Membrane Glycoproteins , Mice , Molecular Weight , Peptide Mapping , Radioligand Assay , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured
14.
Blood ; 91(7): 2397-405, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9516139

ABSTRACT

During development, mice with mutations of stem cell factor (SCF) or its receptor c-kit exhibit defects in melanogenesis, as well as hematopoiesis and gonadogenesis. Consequently, accumulating evidence suggests that the c-kit/SCF system plays a crucial role in all of these processes and in tumors which derive from them. Especially in neuroblastoma (infant tumors of neuroectoderm crest derivation such as melanocytes) it would appear that an autocrine loop exists between c-kit and SCF, and that the functional block of the c-kit receptors with monoclonal antibodies (MoAbs) results in a significant decrease in cellular proliferation. We studied the expression and role of c-kit and SCF in cell lines of soft tissue sarcoma of neuroectodermic origin, such as Ewing's sarcoma (ES) and peripheral neuro-ectodermal tumors (PNET). Using flow cytometry with MoAb CD117 PE, c-kit expression was highlighted in all six of the cell lines examined. This receptor was specifically and functionally activated by SCF, as shown by the binding experiments and the intracellular phosphotyrosine and immunoprecipitation studies that were performed. Using reverse transcriptase polymerase chain reaction analysis, five of the six cellular lines expressed the mRNA of SCF. In the medium measured by using an enzyme- linked immunosorbent assay, low concentrations of SCF were found: only the TC32 cellular line produced significantly higher levels (32 pg) than control. In serum-free culture the addition of SCF reduced the percentage of apoptotic cells from 25% to 90% in five out of the six cellular lines. This observation was confirmed by (1) the functional block of c-kit with MoAb: after 7 days of culture more than 30% of the cells were apoptotic (range 31.5% to 100%) in five out of six cell lines and there was also a decrease in the percentage of cells in phase S, and (2) c-kit antisense oligonucleotides: in the cellular lines treated with oligonucleotides (in relation to the untreated lines) there was a notable reduction (P < .001) both in the absolute number of cells and the 3H-thymidine uptake. These results indicate that ES and PNET express c-kit and its ligand SCF and that SCF is capable of protecting the tumor cells against apoptosis. Furthermore, the reverse transcriptase-polymerase chain reaction performed on the biopsies revealed the presence of mRNA both of SCF and c-kit in practically all of the samples studied. Our in vitro data lead us to assume that SCF may also inhibit tumor cell apoptosis in vivo.


Subject(s)
Apoptosis , Bone Neoplasms/pathology , Neuroectodermal Tumors, Primitive, Peripheral/pathology , Proto-Oncogene Proteins c-kit/metabolism , Sarcoma, Ewing/pathology , Sarcoma/pathology , Stem Cell Factor/metabolism , Animals , Bone Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Neuroectodermal Tumors, Primitive, Peripheral/metabolism , Oligonucleotides, Antisense , Proto-Oncogene Proteins c-kit/genetics , Sarcoma/metabolism , Sarcoma, Ewing/metabolism , Stem Cell Factor/genetics , Tumor Cells, Cultured
15.
Acta bioquím. clín. latinoam ; 31(1): 3-12, mar. 1997. ilus, tab
Article in Spanish | LILACS | ID: lil-207549

ABSTRACT

El anticuerpo monoclonal (AcMo) anti-CEA B2C114 fue marcado con 99mTc utilizando uno de los métodos directos de marcación, en el cual la proteína (AcMo IgG1) se reuce utilizando una solución de Ditiotreitol (DTT) para generar grupos sulfhidrilos, uniendo el 99mTc a los mismos a través del anión [99mTcNCl4]- preparado por calentamiento a reflujo de pertecneciato en presencia de azida sódica y HCl conc. El exceso de DTT se eliminó pasando el AcMo reducido por Sephadex G-25 y del mismo modo se realizó la purificación del anticuerpo marcado. Las cromatografías en TLC y Whatman Nº1 usando Metanol 85 por ciento y solución fisiológica como solventes, mostraron una actividad unida a proteínas entre 55-60 por ciento, obteniéndose luego de la purificación por Sephadex G-25 un producto con 90-95 por ciento de pureza radioquímica. Se realizó la biodistribución en ratones Balb/c a las 21 h post inyección obteniéndose una relación de 2:1 de la dosis inyectada por gramo de tejido, con respecto al tumor reactivo:no reactivo. El ensayo de unión realizado demostró que el AcMo conservó su actividad biológica después de marcado


Subject(s)
Animals , Mice , Antibodies, Monoclonal/pharmacokinetics , Isotope Labeling , Models, Biological , Sodium Pertechnetate Tc 99m , Antibodies, Monoclonal , Carcinoembryonic Antigen , Biological Availability , Chromatography, Gel , Chromatography, Paper , Chromatography, Thin Layer , Isotope Labeling/statistics & numerical data , Mice, Inbred BALB C , Neoplasms/diagnosis
16.
Acta bioquím. clín. latinoam ; 31(1): 3-12, mar. 1997. ilus, tab
Article in Spanish | BINACIS | ID: bin-19578

ABSTRACT

El anticuerpo monoclonal (AcMo) anti-CEA B2C114 fue marcado con 99mTc utilizando uno de los métodos directos de marcación, en el cual la proteína (AcMo IgG1) se reuce utilizando una solución de Ditiotreitol (DTT) para generar grupos sulfhidrilos, uniendo el 99mTc a los mismos a través del anión [99mTcNCl4]- preparado por calentamiento a reflujo de pertecneciato en presencia de azida sódica y HCl conc. El exceso de DTT se eliminó pasando el AcMo reducido por Sephadex G-25 y del mismo modo se realizó la purificación del anticuerpo marcado. Las cromatografías en TLC y Whatman Nº1 usando Metanol 85 por ciento y solución fisiológica como solventes, mostraron una actividad unida a proteínas entre 55-60 por ciento, obteniéndose luego de la purificación por Sephadex G-25 un producto con 90-95 por ciento de pureza radioquímica. Se rea


Subject(s)
Animals , Mice , Isotope Labeling/methods , Sodium Pertechnetate Tc 99m/diagnosis , Antibodies, Monoclonal/pharmacokinetics , Models, Biological , Carcinoembryonic Antigen/diagnosis , Antibodies, Monoclonal/diagnosis , Neoplasms/diagnosis , Chromatography, Thin Layer/statistics & numerical data , Chromatography, Gel , Chromatography, Paper , Mice, Inbred BALB C , Isotope Labeling/statistics & numerical data , Biological Availability
17.
Int Immunol ; 8(11): 1643-50, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8943558

ABSTRACT

Human CD38 is a transmembrane glycoprotein involved in lymphocyte activation and adhesion to endothelium. The ectocellular domain of the molecule possesses properties of a bifunctional enzyme catalyzing both the synthesis from NAD+ and the hydrolysis of the calcium-releasing metabolite cyclic ADP-ribose (cADPR). Surface expression of CD38 (mCD38) is rapidly and almost completely down-modulated upon ligation by specific mAb in cells from different lineages. The data presented here also show that, in addition to the existence of a mCD38, a soluble form of CD38 (sCD38) is detectable in the cell culture supernatant of allo-activated T lymphocytes and of several tumor cell lines. sCD38 is also present in vivo and is assayable in normal (fetal serum and amniotic fluid) and pathological (serum and ascites from patients with multiple myeloma, and serum from patients with AIDS) biological fluids. Immunoaffinity chromatography, SDS-PAGE and Western blot analyses with mAb and polyclonal antibodies, along with metabolic labeling, yield a body of data concerning the structure of sCD38, which displays a M(r) of 39 kDa. Native sCD38 maintains the ability to inhibit the binding activity of different anti-CD38 mAb and still catalyzes the synthesis and the hydrolysis of cADPR at the same ratio observed with mCD38. Furthermore, cross-linking experiments indicate that the purified soluble molecule binds a 120 kDa molecule expressed by monocytoid cells and identified as a candidate ligand for human mCD38.


Subject(s)
Antigens, Differentiation/analysis , Antigens, Differentiation/immunology , Ascitic Fluid/immunology , Body Fluids/immunology , N-Glycosyl Hydrolases/analysis , N-Glycosyl Hydrolases/immunology , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Amniotic Fluid/immunology , Antibodies, Monoclonal/immunology , Antigens, CD/analysis , Antigens, CD/immunology , Humans , Ligands , Membrane Glycoproteins , Solubility , Tumor Cells, Cultured
19.
J Immunol ; 156(2): 727-34, 1996 Jan 15.
Article in English | MEDLINE | ID: mdl-8543826

ABSTRACT

Human CD38, a pleiotropic molecule with ADP-ribosyl cyclase activity, regulates activation and growth of several cell types. Its in vivo function is incompletely determined, mainly due to the lack of evidence concerning the existence of a single or multiple ligands. We recently observed that CD38 rules a selectin-type adhesion between lymphoid cells and HUVECs. A panel of murine mAbs raised against HUVEC included one (Moon-1) constantly blocking the CD38-mediated adhesion of several cell lines to HUVEC. Tissue distribution studies and an extended immunohistochemical analysis on the majority of normal human tissues revealed that the Moon-1 molecule displays a unique pattern of expression, being present at high levels on resting and activated vascular endothelium, on the majority of monocytes, platelets, NK cells, and to a lesser extent on T, B, and myeloid cells. The Moon-1 structure of an apparent molecular mass of 120 kDa proved to be a ligand for human CD38, as inferred by the direct binding observed when using a chimeric mouse CD8 alpha-human CD38 (mCD8 alpha-hCD38) molecule as a probe in Western blot experiments. Furthermore, Ab-induced modulation experiments highlighted an association between the Moon-1 molecule and human CD38 on the surface of cell lines coexpressing the two structures, which also share a common regulation system of surface expression. Finally, direct ligation of Moon-1 on T cell lines caused a relevant increase in the cytoplasmic concentration of calcium ([Ca2+]i).


Subject(s)
Antigens, CD , Antigens, Differentiation/metabolism , Carrier Proteins/physiology , Endothelium, Vascular/metabolism , N-Glycosyl Hydrolases/metabolism , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Animals , Antibodies, Monoclonal/immunology , Blood Cells/metabolism , Carrier Proteins/biosynthesis , Carrier Proteins/immunology , Carrier Proteins/isolation & purification , Cell Adhesion , Gene Expression Regulation , Humans , Leukemia-Lymphoma, Adult T-Cell , Lymph Nodes/blood supply , Membrane Glycoproteins , Mice , Molecular Weight , Organ Specificity , Recombinant Fusion Proteins/metabolism , Signal Transduction , Tumor Cells, Cultured , Umbilical Veins
20.
Nucl Med Biol ; 22(3): 367-72, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7627152

ABSTRACT

The anti-carcinoembryonic B2C114 monoclonal antibody was radiolabeled with 99mTc by a direct method and quality control tested in vitro by instant thin layer chromatography, gel column scanning and cellulose acetate electrophoresis and assessed in vivo for radioimmunodetection on a murine spontaneous mammary carcinoma. The optimal results of percent 99mTc bound to protein were obtained at a dithiothreitol:antibody molar ratio ranging from 800:1 to 1000:1 and at a methylene diphosphonate:stannous fluoride weight ratio of 4.3:1. Although cysteine removed up to 18% of the label during the first 4 h, the stability of the tracer appeared to be excellent in human serum at 37 degrees C and when challenged with DTPA. 99mTc-labeled B2C114 demonstrated good and specific in vivo tumor targeting.


Subject(s)
Antibodies, Monoclonal , Isotope Labeling/standards , Mammary Neoplasms, Experimental/diagnostic imaging , Organotechnetium Compounds , Animals , Antibodies, Monoclonal/pharmacokinetics , Carcinoembryonic Antigen/immunology , Humans , Isotope Labeling/methods , Mammary Neoplasms, Experimental/diagnosis , Mice , Mice, Inbred BALB C , Quality Control , Radionuclide Imaging , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL