Macrophage Activating Factor

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 4053 Experts worldwide ranked by ideXlab platform

Nobuto Yamamoto - One of the best experts on this subject based on the ideXlab platform.

  • inhibitory effect of vitamin d binding protein derived Macrophage Activating Factor on dmba induced hamster cheek pouch carcinogenesis and its derived carcinoma cell line
    Oncology Letters, 2011
    Co-Authors: Yukiyo Toyohara, Nobuto Yamamoto, Susumu Hashitani, Hiromitsu Kishimoto, Kazuma Noguchi, Masahiro Urade
    Abstract:

    This study investigated the inhibitory effect of vitamin D-binding protein-derived Macrophage-Activating Factor (GcMAF) on carcinogenesis and tumor growth, using a 9,10-dimethyl-1,2-benzanthracene (DMBA)-induced hamster cheek pouch carcinogenesis model, as well as the cytocidal effect of activated Macrophages against HCPC-1, a cell line established from DMBA-induced cheek pouch carcinoma. DMBA application induced squamous cell carcinoma in all 15 hamsters of the control group at approximately 10 weeks, and all 15 hamsters died of tumor burden within 20 weeks. By contrast, 2 out of the 14 hamsters with GcMAF administration did not develop tumors and the remaining 12 hamsters showed a significant delay of tumor development for approximately 3.5 weeks. The growth of tumors formed was significantly suppressed and none of the hamsters died within the 20 weeks during which they were observed. When GcMAF administration was stopped at the 13th week of the experiment in 4 out of the 14 hamsters in the GcMAF-treated group, tumor growth was promoted, but none of the mice died within the 20-week period. On the other hand, when GcMAF administration was commenced after the 13th week in 5 out of the 15 hamsters in the control group, tumor growth was slightly suppressed and all 15 hamsters died of tumor burden. However, the mean survival time was significantly extended. GcMAF treatment activated peritoneal Macrophages in vitro and in vivo, and these activated Macrophages exhibited a marked cytocidal effect on HCPC-1 cells. Furthermore, the cytocidal effect of activated Macrophages was enhanced by the addition of tumor-bearing hamster serum. These findings indicated that GcMAF possesses an inhibitory effect on tumor development and growth in a DMBA-induced hamster cheek pouch carcinogenesis model.

  • abstract 5531 administration of the most potent Macrophage Activating Factor gcmaf to cancer patients eradicates a variety of cancers indiscriminately
    Cancer Research, 2011
    Co-Authors: Nobuto Yamamoto, Masumi Ueda, Kazuya Hashinaka
    Abstract:

    Intratumor BCG administration eradicates local as well as metastasized tumors (e.g., skin cancer). Administration of BCG into noncancerous tissues, however, results in no effect on the tumors. Inflammation induced by BCG in normal tissues releases lysophospholipids that activate Macrophages. Because cancerous tissues contain alkylphospholipids, BCG-induced inflammation of cancerous tissues produces alkyl-lysophospholipids and alkylglycerols that activate Macrophages approximately 400 times more effective than lysophospholipids, implying that highly activated Macrophages are tumoricidal. Inflammation-primed Macrophage activation is the principal Macrophage activation process that requires serum Gc protein (known as vitamin D-binding protein) and participation of B and T lymphocytes. A trisaccharide composed of N-acetylgalactosamine with dibranched galactose and sialic acid termini at 420 threonine residue of Gc protein is hydrolyzed by the β-galactosidase (Bgl) of inflammation-primed B cells and the Neu-1 sialidase of T cells to yield the Macrophage Activating Factor (MAF), the protein with N-acetylgalactoamine as the remaining sugar. Thus, Gc protein is the precursor for the principal MAF. However, the MAF precursor activity of serum Gc protein of cancer patients was lost or reduced because Gc protein is deglycosylated by serum α-N-acetylgalactosaminidase (Nagalase) secreted from cancerous cells. Thus, serum Nagalase activity is proportional to tumor burden and serves as an excellent prognostic index. Deglycosylated serum Gc protein can not be converted to MAF, leading to immunosuppression. Stepwise treatment of purified serum Gc protein with immobilized β-galactosidase and sialidase generates probably the most potent MAF (termed GcMAF) ever discoverd that produces no side effect in humans. Intramuscular administration of 100 ng GcMAF activates systemic Macrophages at maximal level with 100-fold increased ingestion index and 30-fold increased superoxide generating capacity in 6 hrs. These activated Macrophages developed an enormous variation of receptors that recognize cell surface abnormality (known as tumor associated antigen, TAA) of a variety of cancer cells and become tumoricidal to a variety of cancers indiscriminately. When those cancer patients bearing breast, prostate, colorectal, stomach, liver, lung, esophagus, kidney, bladder, uterus or ovarian cancer, or melanoma, fibrosarcoma, leukemia, glioblastoma or mesothelioma were intramuscularly administered with 100 ng GcMAF/week, their tumors were eradicated in 16 – 54 weeks which is roughly proportional to their original tumor burden. The curative rate of adenocarcinomas is a biphasic pattern due to a mixture of highly undifferentiated and differentiated tumor cells while the curative rate of all other cancer types is linear. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 5531. doi:10.1158/1538-7445.AM2011-5531

  • abstract 5532 immunotherapy of breast cancer with gc protein derived Macrophage Activating Factor gcmaf
    Cancer Research, 2011
    Co-Authors: Nobuto Yamamoto, Masumi Ueda, Kazuya Hashinaka
    Abstract:

    Serum Gc protein (known as vitamin D-binding protein) is the precursor for the principal Macrophage Activating Factor (MAF). The MAF precursor activity of serum Gc protein of cancer patients was lost or reduced because Gc protein was deglycosylated by serum α-N-acetylgalactosaminidase (Nagalase) secreted from cancerous cells. Thus, serum Nagalase activity is directly proportional to tumor burden. Macrophages of cancer patients having deglycosylated Gc protein cannot be activated. Since Macrophage activation for phagocytosis and antigen-presentation to B cells and T cells is the first indispensable step for development of both humoral and cellular immunities, lack of Macrophage activation leads to immunosuppression. Advanced cancer patients have high serum Nagalase activity, resulting in no Macrophage activation and severe immunosuppression that explain why cancer patients die with overwhelming infection (e.g., pneumonia). Stepwise treatment of purified Gc protein with immobilized β-galactosidase and sialidase generated probably the most potent Macrophage Activating Factor (termed GcMAF) ever discovered. Because GcMAF is structurally identical to the native human MAF, GcMAF produces no side effect in humans. Macrophages activated by GcMAF develop an enormous variation of receptors that recognize the abnormality in malignant cell surface and are highly tumoricidal to a variety of cancers indiscriminately. When human Macrophages were treated in vitro with GcMAF (100 picogram/ml) for 3 hr and a breast cancer cell line MCF-7 was added with effector/target ratio of 1.5, 60% and 86% of MCF-7 cells were killed in 4 hr and 18 hr incubation, respectively. Administration of 100 nanogram (ng) GcMAF per human results in the maximal level of Macrophage activation. Because the activated state of Macrophages is approximately 6 days, sixteen breast cancer patients received weekly administration of 100 ng GcMAF. They regressed in biphasic pattern due to the mixed population of undifferentiated and differentiated cells. After 16-23 weekly administrations of GcMAF (100 ng/week), all sixteen patients had very low serum Nagalase levels equivalent to those of healthy control values, indicating that these patients are tumor free. No recurrence occurred for more than ten years. GcMAF also has a potent mitogenic capacity to act on the myeloid progenitor cells, resulting in a 40-fold increase in systemic Macrophage cell counts in 4 days. Such highly activated systemic Macrophages are chemotactically recruited to inflamed lesions by 180-fold increase of Macrophage cell counts. Since intravenous administration of GcMAF allows a rapid interaction of GcMAF with myeloid progenitor cells in bone marrow, the systemic cell counts of the activated Macrophages increased to more than 100-fold in 2 days. Weekly intravenous administration of 100 ng GcMAF to metastatic breast cancer patients (n=8) eradicates tumors in 7-15 weeks. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 5532. doi:10.1158/1538-7445.AM2011-5532

  • immunotherapy of hiv infected patients with gc protein derived Macrophage Activating Factor gcmaf
    Journal of Medical Virology, 2009
    Co-Authors: Nobuto Yamamoto, Naofumi Ushijima, Yoshihiko Koga
    Abstract:

    The above article, published online on 21 Nov 2008 Wiley Online Library (wileyonlinelibrary.com), has been retracted by agreement between Dr. Ari Zuckerman, Editor-in-Chief, Journal of Medical Virology and Wiley Periodicals, Inc. due to irregularities in the documentation for institutional review board approval.

  • immunotherapy for prostate cancer with gc protein derived Macrophage Activating Factor gcmaf
    Translational Oncology, 2008
    Co-Authors: Nobuto Yamamoto, Hirofumi Suyama, Nobuyuki Yamamoto
    Abstract:

    Abstract Serum Gc protein (known as vitamin D3-binding protein) is the precursor for the principal Macrophage-Activating Factor (MAF). The MAF precursor activity of serum Gc protein of prostate cancer patients was lost or reduced because Gc protein was deglycosylated by serum a-N-acetylgalactosaminidase (Nagalase) secreted from cancerous cells. Therefore, Macrophages of prostate cancer patients having deglycosylated Gc protein cannot be activated, leading to immunosuppression. Stepwise treatment of purified Gc protein with immobilized â-galactosidase and sialidase generated the most potent MAF (termed GcMAF) ever discovered, which produces no adverse effect in humans. Macrophages activated by GcMAF develop a considerable variation of receptors that recognize the abnormality in malignant cell surface and are highly tumoricidal. Sixteen nonanemic prostate cancer patients received weekly administration of 100 ng of GcMAF. As the MAF precursor activity increased, their serum Nagalase activity decreased. Because serum Nagalase activity is proportional to tumor burden, the entire time course analysis for GcMAF therapy was monitored by measuring the serum Nagalase activity. After 14 to 25 weekly administrations of GcMAF (100 ng/week), all 16 patients had very low serum Nagalase levels equivalent to those of healthy control values, indicating that these patients are tumor-free. No recurrence occurred for 7 years.

Venkateswara R Naraparaju - One of the best experts on this subject based on the ideXlab platform.

  • antitumor effect of vitamin d binding protein derived Macrophage Activating Factor on ehrlich ascites tumor bearing mice
    Experimental Biology and Medicine, 1999
    Co-Authors: Yoshihiko Koga, Venkateswara R Naraparaju, Nobuto Yamamoto
    Abstract:

    Cancerous cells secrete alpha-N-acetylgalactosaminidase (NaGalase) into the blood stream, resulting in deglycosylation of serum vitamin D3-binding protein (known as Gc protein), which is a precursor for Macrophage Activating Factor (MAF). Incubation of Gc protein with immobilized beta-galactosidase and sialidase generates the most potent Macrophage Activating Factor (designated GcMAF). Administration of GcMAF to cancer-bearing hosts can bypass the inactivated MAF precursor and act directly on Macrophages for efficient activation. Therapeutic effects of GcMAF on Ehrlich ascites tumor-bearing mice were assessed by survival time and serum NaGalase activity, because serum NaGalase activity was proportional to tumor burden. A single administration of GcMAF (100 pg/mouse) to eight mice on the same day after transplantation of the tumor (5 x 10(5) cells) showed a mean survival time of 21 +/- 3 days for seven mice, with one mouse surviving more than 60 days, whereas tumor-bearing controls had a mean survival time of 13 +/- 2 days. Six of the eight mice that received two GcMAF administrations, at Day 0 and Day 4 after transplantation, survived up to 31 +/- 4 days whereas, the remaining two mice survived for more than 60 days. Further, six of the eight mice that received three GcMAF administrations with 4-day intervals showed an extended survival of at least 60 days, and serum NaGalase levels were as low as those of control mice throughout the survival period. The cure with subthreshold GcMAF-treatments (administered once or twice) of tumor-bearing mice appeared to be a consequence of sustained Macrophage activation by inflammation resulting from the Macrophage-mediated tumoricidal process. Therefore, a protracted Macrophage activation induced by a few administrations of minute amounts of GcMAF eradicated the murine ascites tumor.

  • structurally well defined Macrophage Activating Factor derived from vitamin d3 binding protein has a potent adjuvant activity for immunization
    Immunology and Cell Biology, 1998
    Co-Authors: Nobuto Yamamoto, Venkateswara R Naraparaju
    Abstract:

    Freund's adjuvant produced severe inflammation that augments development of antibodies. Thus, mixed administration of antigens with adjuvant was not required as long as inflammation was induced in the hosts. Since Macrophage activation for phagocytosis and antigen processing is the first step of antibody development, inflammation-primed Macrophage activation plays a major role in immune development. Therefore, Macrophage Activating Factor should act as an adjuvant for immunization. The inflammation-primed Macrophage activation process is the major Macrophage Activating cascade that requires participation of serum vitamin D3-binding protein (DBP; human DBP is known as Gc protein) and glycosidases of B and T lymphocytes. Stepwise incubation of Gc protein with immobilized beta-galactosidase and sialidase efficiently generated the most potent Macrophage Activating Factor (designated GcMAF) we have ever encountered. Administration of GcMAF (20 or 100 pg/mouse) resulted in stimulation of the progenitor cells for extensive mitogenesis and activation of Macrophages. Administration of GcMAF (100 pg/mouse) along with immunization of mice with sheep red blood cells (SRBC) produced a large number of anti-SRBC antibody secreting splenic cells in 2-4 days. Thus, GcMAF has a potent adjuvant activity for immunization. Although malignant tumours are poorly immunogenic, 4 days after GcMAF-primed immunization of mice with heat-killed Ehrlich ascites tumour cells, the ascites tumour was no longer transplantable in these mice.

  • immunotherapy of balb c mice bearing ehrlich ascites tumor with vitamin d binding protein derived Macrophage Activating Factor
    Cancer Research, 1997
    Co-Authors: Nobuto Yamamoto, Venkateswara R Naraparaju
    Abstract:

    Abstract Vitamin D3-binding protein (DBP; human DBP is known as Gc protein) is the precursor of Macrophage Activating Factor (MAF). Treatment of mouse DBP with immobilized β-galactosidase or treatment of human Gc protein with immobilized β-galactosidase and sialidase generated a remarkably potent MAF, termed DBPMAF or GcMAF, respectively. The domain of Gc protein responsible for Macrophage activation was cloned and enzymatically converted to the cloned MAF, designated CdMAF. In Ehrlich ascites tumor-bearing mice, tumor-specific serum α-N-acetylgalactosaminidase (NaGalase) activity increased linearly with time as the transplanted tumor cells grew in the peritoneal cavity. Therapeutic effects of DBPMAF, GcMAF, and CdMAF on mice bearing Ehrlich ascites tumor were assessed by survival time, the total tumor cell count in the peritoneal cavity, and serum NaGalase activity. Mice that received a single administration of DBPMAF or GcMAF (100 pg/mouse) on the same day after transplantation of tumor (1 × 105 cells) showed a mean survival time of 35 ± 4 days, whereas tumor-bearing controls had a mean survival time of 16 ± 2 days. When mice received the second DBPMAF or GcMAF administration at day 4, they survived more than 50 days. Mice that received two DBPMAF administrations, at days 4 and 8 after transplantation of 1 × 105 tumor cells, survived up to 32 ± 4 days. At day 4 posttransplantation, the total tumor cell count in the peritoneal cavity was approximately 5 × 105 cells. Mice that received two DBPMAF administrations, at days 0 and 4 after transplantation of 5 × 105 tumor cells, also survived up to 32 ± 4 days, while control mice that received the 5 × 105 ascites tumor cells only survived for 14 ± 2 days. Four DBPMAF, GcMAF, or CdMAF administrations to mice transplanted with 5 × 105 Ehrlich ascites tumor cells with 4-day intervals showed an extended survival of at least 90 days and an insignificantly low serum NaGalase level between days 30 and 90.

  • vitamin d3 binding protein as a precursor for Macrophage Activating Factor in the inflammation primed Macrophage activation cascade in rats
    Cellular Immunology, 1996
    Co-Authors: Nobuto Yamamoto, Venkateswara R Naraparaju
    Abstract:

    When rat peritoneal nonadherent cells were treated with inflammatory lipid metabolites and cultured with adherent cells in 1% fetal calf serum (FCS) supplemented medium RPMI 1640 (FCS medium) for 3 hr, markedly enhanced phagocytic and superoxide generating capacities of Macrophages were observed. Stepwise preparation of conditioned medium of lysophosphatidylcholine (lyso-Pc)-treated B cells and untreated T cells in FCS medium generated a potent Macrophage Activating Factor whereas cultivation of lyso-Pc-treated B cells alone in a 1% adult rat serum supplemented medium efficiently generated the Macrophage Activating Factor. Generation of Macrophage Activating Factor requires a precursor protein, serum vitamin D3-binding protein (DBP), as well as participation of lymphocyte glycosidases. The lyso-Pc-inducible beta-galactosidase of B lymphocytes and the Neu-1 sialidase of T lymphocytes modified bovine DBP (bDBP) to yield the Macrophage Activating Factor, a protein with N-acetylgalactosamine as the remaining sugar. In contrast, lyso-Pc-inducible beta-galactosidase of B cells alone modified rat DBP (rDBP) to yield the Macrophage Activating Factor, a protein with N-acetylgalactosamine as the remaining sugar. Thus, we conclude that bDBP carries a trisaccharide composed of N-acetylgalactosamine, galactose, and sialic acid while rDBP carries a disaccharide composed of N-acetylgalactosamine and galactose.

C J Secombes - One of the best experts on this subject based on the ideXlab platform.

  • antigen induced release of Macrophage Activating Factor from rainbow trout oncorhynchus mykiss leucocytes
    Veterinary Immunology and Immunopathology, 1994
    Co-Authors: M J Marsden, David Cox, C J Secombes
    Abstract:

    Abstract The production of Macrophage-Activating Factor (MAF) by rainbow trout, Oncorhynchus mykiss, head kidney leucocytes at varying times post-immunisation, with the fish bacterial pathogen, Aeromonas salmonicida, was investigated and correlated with head kidney lymphocyte proliferation and serum antibody production. MAF production was preceded by lymphocyte proliferation and both responses were highest using whole bacterial cells as the in vitro stimulant. MAF production and antibody production increased 2–3 weeks post-immunisation, and peaked 4–5 weeks post-immunisation. The relative importance of MAF activated phagocytes in the immunological armoury of disease-resistant, vaccinated fish requires further investigation.

  • effect of temperature on Macrophage activation and the production of Macrophage Activating Factor by rainbow trout oncorhynchus mykiss leucocytes
    Developmental and Comparative Immunology, 1994
    Co-Authors: Laura J Hardie, T C Fletcher, C J Secombes
    Abstract:

    Abstract Production of Macrophage Activating Factor (MAF) by rainbow trout leucocytes has been shown to be temperature dependent in vivo and in vitro. Cells from fish held at 14°C and stimulated to produce MAF immediately after isolation were capable of secreting MAF down to 6°C (the lowest temperature tested). However, after 48 h at 6°C, these leucocytes show impaired MAF secretion. Acclimation of fish to low temperatures (7°C) did not recover the inhibitory effects of low in vitro temperatures on MAF production, but if these leucocytes were preincubated at 10 or 18°C for 48 h, MAF was produced from these cells. Interestingly, Macrophages isolated from fish kept at 7 or 14°C and cultured at low temperatures (6°C) were responsive to MAF-containing supernatants, and showed a higher relative increase in respiratory burst activity compared with their counterparts cultured at 10 and 18°C. Such observations clearly demonstrate that a major impairment of bactericidal activity at low temperatures resides within the specific immune compartment of fish. The implications for fish health are discussed.

P Deschaux - One of the best experts on this subject based on the ideXlab platform.

  • Possible implication of Macrophages in the regulation of cytochrome P450 activities in carp (Cyprinus carpio)
    Fish and Shellfish Immunology, 2006
    Co-Authors: D. Marionnet, P Deschaux, S. Reynaud
    Abstract:

    Macrophages play a key role in the regulation of cytochrome P450 activity induced by immunostimulants in mammals. We investigated the effects of immunostimulants (LPS, dextran sulfate and tilorone) on biotransformation and Macrophage activities in carp. The major effect of LPS was its capacity to inhibit 3-MC-induced cytochrome P450 activities in the liver and head kidney. Basal phase I activities were reduced by tilorone and dextran sulfate in immune organs. Tilorone and dextran sulfate differently modulated total cytochrome P450 contents and P4501A activities suggesting differential sensitivity for P450 classes. In immune organs, tilorone and dextran sulfate inhibited basal EROD activity. Tilorone inhibited 3-MC-induced EROD activity whereas dextran sulfate enhanced this activity. LPS and dextran sulfate increased ROS production by Macrophages and all the immunostimulants induced Macrophage Activating Factor (MAF) production. This study demonstrates for the first time in fish the capacity of CYP-regulated immunostimulants to activate Macrophages and provides initial insight into the capacity of Macrophages to regulate CYP activity induced by immunostimulants in fish. (c) 2005 Elsevier Ltd. All rights reserved.

  • lindane increases Macrophage Activating Factor production and intracellular calcium in rainbow trout oncorhynchus mykiss leukocytes
    Ecotoxicology and Environmental Safety, 2002
    Co-Authors: C Duchiron, S Betoulle, Stephane Reynaud, P Deschaux
    Abstract:

    Abstract The authors studied the in vitro effects of lindane on Macrophage-Activating Factor (MAF) production by peripheral blood leukocytes (PBLs) in rainbow trout. MAF production by PBLs induced normally by mitogens concanavalin A (ConA) and phorbol myristate acetate (PMA) was not modified by a pretreatment with lindane (from 2.5 to 50 μM ). Only a concentration of 100 μM lindane decreased MAF production, associated with cellular death. Moreover, MAF activities were detected in supernatants of PBL cultures treated with lindane from 2.5 to 10 μM in absence of ConA/PMA stimulation. Factors present in these supernatants remain to be identified. Lindane, at concentrations which did not induce MAF production (50 and 100 μM ) led to an increase in PBL calcium levels by acting on the endoplasmic reticulum calcium stores. Although the intracytosolic calcium concentration ([Ca2+]i) increase seems to be associated with cell death, lindane-induced MAF production may be linked with other intracellular mechanisms.

  • lindane induced Macrophage Activating Factor maf production by peripheral blood leukocytes pbls of rainbow trout oncorhynchus mykiss involvement of intracellular camp mobilization
    Aquatic Toxicology, 2002
    Co-Authors: C Duchiron, Stephane Reynaud, P Deschaux
    Abstract:

    Abstract We studied the in vitro effects of the insecticide lindane (2.5–50 μM) on Macrophage Activating Factor (MAF) production by the peripheral blood leukocytes (PBLs) in rainbow trout. The MAF production induced by the mitogens concanavalin A (ConA) and phorbol-12-myristate-13-acetate (PMA) was not modified by lindane pre-treatment. But lindane alone (2.5–25 μM) stimulated the secretion of MAF by PBLs. Intracellular calcium levels ([Ca 2+ ] i ) was measured over 6 min by spectrofluorimetry using Indo-1/AM as fluorescent probe. Lindane (25–100 μM) significantly increased the [Ca 2+ ] i in PBLs, but had no effect on calcium at the dose that caused MAF secretion. Moreover, the effect of lindane on MAF production was potentiated by the inhibitor of phosphodiesterase, isobutylmethylxanthin (IBMX). Lindane also directly increased adenosine monophosphate cyclic (cAMP) in PBLs over the same concentration range that it stimulated MAF production by PBLs. Taken together, these results suggest that lindane increase MAF production by acting on intracellular cAMP concentrations. Moreover, the capacity of this insecticide to act on the [Ca 2+ ] i or on the intracellular concentrations of cAMP according to the dose used could possibly explain its contradictory effects earlier observed on immunity.

Stefania Pacini - One of the best experts on this subject based on the ideXlab platform.

  • significance of hydrophobic and charged sequence similarities in sodium bile acid cotransporter and vitamin d binding protein Macrophage Activating Factor
    bioRxiv, 2020
    Co-Authors: Ibne Raihan Zunaid, Stefania Pacini, Marco Ruggiero
    Abstract:

    Abstract Sodium-bile acid cotransporter, also denominated sodium-taurocholate cotransporting polypeptide (NTCP) is an integral membrane protein with multiple hydrophobic transmembrane domains. The third extracellular domain of NTCP presents a stretch of nine aminoacids (KGIVISLVL) that is characterized by pronounced hydrophobicity and serves as receptor for a protein, preS1, showing the hydrophobic epta-peptide sequence NPLGFFP. Vitamin D-binding protein Macrophage Activating Factor (DBP-MAF) is a multifunctional protein that is characterized by two hydrophobic regions able to bind fatty acids and vitamin D, respectively. Here we demonstrate that NTCP and DBP-MAF show significant sequence similarities as far as hydrophobic stretches of aminoacids are concerned. Alignment of the sequence of seven aminoacids preceding the 157-KGIVISLVL-165 stretch of NTCP shows four aminoacids that are identical to those of the corresponding sequence of DBP-MAF, and two that are conserved substitutions. In addition, in the sequence of DBP-MAF that is aligned with the sequence YKGIVISLVL of NTCP, there are two contiguous negatively charged aminoacids (ED) and, in the preceding epta-peptide sequence, there are three negatively charged aminoacids (D-ED), whereas in the corresponding sequence of NTCP there are only two (D--D) that are not contiguous. This concentration of negatively charged aminoacids may be involved in binding of protein inserts characterized by high density of positively charges residues. The alternating hydrophobic and electrostatic interactions described in this paper may help elucidating the biological roles of these proteins as far as protein-protein interactions are concerned.

  • is chondroitin sulfate responsible for the biological effects attributed to the gc protein derived Macrophage Activating Factor gcmaf
    Medical Hypotheses, 2016
    Co-Authors: Marco Ruggiero, Heinz Reinwald, Stefania Pacini
    Abstract:

    We hypothesize that a plasma glycosaminoglycan, chondroitin sulfate, may be responsible for the biological and clinical effects attributed to the Gc protein-derived Macrophage Activating Factor (GcMAF), a protein that is extracted from human blood. Thus, Gc protein binds chondroitin sulfate on the cell surface and such an interaction may occur also in blood, colostrum and milk. This interpretation would solve the inconsistencies encountered in explaining the effects of GcMAF in vitro and in vivo. According to our model, the Gc protein or the GcMAF bind to chondroitin sulfate both on the cell surface and in bodily fluids, and the resulting multimolecular complexes, under the form of oligomers trigger a transmembrane signal or, alternatively, are internalized and convey the signal directly to the nucleus thus eliciting the diverse biological effects observed for both GcMAF and chondroitin sulfate.

  • effects of oxaliplatin and oleic acid gc protein derived Macrophage Activating Factor on murine and human microglia
    Journal of Neuroscience Research, 2015
    Co-Authors: Jacopo J V Branca, Marco Ruggiero, Gabriele Morucci, Francesca Malentacchi, Stefania Gelmini, Stefania Pacini
    Abstract:

    The biological properties and characteristics of microglia in rodents have been widely described, but little is known about these features in human microglia. Several murine microglial cell lines are used to investigate neurodegenerative and neuroinflammatory conditions; however, the extrapolation of the results to human conditions is frequently met with criticism because of the possibility of species-specific differences. This study compares the effects of oxaliplatin and of oleic acid Gc-protein-derived Macrophage-Activating Factor (OA-GcMAF) on two microglial cell lines, murine BV-2 cells and human C13NJ cells. Cell viability, cAMP levels, microglial activation, and vascular endothelial growth Factor (VEGF) expression were evaluated. Our data demonstrate that oxaliplatin induced a significant decrease in cell viability in BV-2 and in C13NJ cells and that this effect was not reversed with OA-GcMAF treatment. The signal transduction pathway involving cAMP/VEGF was activated after treatment with oxaliplatin and/or OA-GcMAF in both cell lines. OA-GcMAF induced a significant increase in microglia activation, as evidenced by the expression of the B7-2 protein, in BV-2 as well as in C13NJ cells that was not associated with a concomitant increase in cell number. Furthermore, the effects of oxaliplatin and OA-GcMAF on coculture morphology and apoptosis were evaluated. Oxaliplatin-induced cell damage and apoptosis were nearly completely reversed by OA-GcMAF treatment in both BV-2/SH-SY5Y and C13NJ/SH-SY5Y cocultures. Our data show that murine and human microglia share common signal transduction pathways and activation mechanisms, suggesting that the murine BV-2 cell line may represent an excellent model for studying human microglia. © 2015 Wiley Periodicals, Inc.

  • gc protein derived Macrophage Activating Factor counteracts the neuronal damage induced by oxaliplatin
    Anti-Cancer Drugs, 2015
    Co-Authors: Gabriele Morucci, Marco Ruggiero, Jacopo J V Branca, Massimo Gulisano, Ferdinando Paternostro, Alessandra Pacini, Lorenzo Di Cesare Mannelli, Stefania Pacini
    Abstract:

    Oxaliplatin-based regimens are effective in metastasized advanced cancers. However, a major limitation to their widespread use is represented by neurotoxicity that leads to peripheral neuropathy. In this study we evaluated the roles of a proven immunotherapeutic agent [Gc-protein-derived Macrophage Activating Factor (GcMAF)] in preventing or decreasing oxaliplatin-induced neuronal damage and in modulating microglia activation following oxaliplatin-induced damage. The effects of oxaliplatin and of a commercially available formula of GcMAF [oleic acid-GcMAF (OA-GcMAF)] were studied in human neurons (SH-SY5Y cells) and in human microglial cells (C13NJ). Cell density, morphology and viability, as well as production of cAMP and expression of vascular endothelial growth Factor (VEGF), markers of neuron regeneration [neuromodulin or growth associated protein-43 (Gap-43)] and markers of microglia activation [ionized calcium binding adaptor molecule 1 (Iba1) and B7-2], were determined. OA-GcMAF reverted the damage inflicted by oxaliplatin on human neurons and preserved their viability. The neuroprotective effect was accompanied by increased intracellular cAMP production, as well as by increased expression of VEGF and neuromodulin. OA-GcMAF did not revert the effects of oxaliplatin on microglial cell viability. However, it increased microglial activation following oxaliplatin-induced damage, resulting in an increased expression of the markers Iba1 and B7-2 without any concomitant increase in cell number. When neurons and microglial cells were co-cultured, the presence of OA-GcMAF significantly counteracted the toxic effects of oxaliplatin. Our results demonstrate that OA-GcMAF, already used in the immunotherapy of advanced cancers, may significantly contribute to neutralizing the neurotoxicity induced by oxaliplatin, at the same time possibly concurring to an integrated anticancer effect. The association between these two powerful anticancer molecules would probably produce the dual effect of reduction of oxaliplatin-induced neurotoxicity, together with possible synergism in the overall anticancer effect.

  • gc protein derived Macrophage Activating Factor decreases α n acetylgalactosaminidase levels in advanced cancer patients
    OncoImmunology, 2013
    Co-Authors: Lynda Thyer, Gabriele Morucci, Jacopo J V Branca, Massimo Gulisano, Emma Ward, Rodney Smith, David Noakes, Robert Eslinger, Stefania Pacini
    Abstract:

    α-N-acetylgalactosaminidase (nagalase) accumulates in the serum of cancer patients and its activity correlates with tumor burden, aggressiveness and clinical disease progression. The administration of gc protein-derived MacrophageActivating Factor ( gcMAF) to cancer patients with elevated levels of nagalase has been associated with a decrease of serum nagalase activity and with significant clinical benefits. here, we report the results of the administration of gcMAF to a heterogeneous cohort of patients with histologically diverse, advanced neoplasms, generally considered as “incurable” diseases. In most cases, g cMAF therapy was initiated at late stages of tumor progression. As this is an open-label, noncontrolled, retrospective analysis, caution must be employed when establishing cause-effect relationships between the administration g cMAF and disease outcome. h owever, the response to g cMAF was generally robust and some trends emerged. All patients (n = 20) presented with elevated serum nagalase activity, well above normal values. All patients but one showed a significant decrease of serum nagalase activity upon weekly gcMAF injections. Decreased nagalase activity was associated with improved clinical conditions and no adverse side effects were reported. The observations reported here confirm and extend previous results and pave the way to further studies aimed at assessing the precise role and indications for g cMAF-based anticancer immunotherapy.