Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Tumour Biol ; 37(9): 12697-12711, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27448301

ABSTRACT

The carboxy-terminal third domain of alpha-fetoprotein (AFP-3D) is known to harbor binding and/or interaction sites for hydrophobic ligands, receptors, and binding proteins. Such reports have established that AFP-3D consists of amino acid (AA) sequence stretches on the AFP polypeptide that engages in protein-to-protein interactions with various ligands and receptors. Using a computer software program specifically designed for such interactions, the present report identified AA sequence fragments on AFP-3D that could potentially interact with a variety of cell cycle proteins. The cell cycle proteins identified were (1) cyclins, (2) cyclin-dependent kinases, (3) cell cycle-associated proteins (inhibitors, checkpoints, initiators), and (4) ubiquitin ligases. Following detection of the AFP-3D to cell cycle protein interaction sites, the computer-derived AFP localization AA sequences were compared and aligned with previously reported hydrophobic ligand and receptor interaction sites on AFP-3D. A literature survey of the association of cell cycle proteins with AFP showed both positive relationships and correlations. Previous reports of experimental AFP-derived peptides effects on various cell cycle proteins served to confirm and verify the present computer cell cycle protein identifications. Cell cycle protein interactions with AFP-CD peptides have been reported in cultured MCF-7 breast cancer cells subjected to mRNA microarray analysis. After 7 days in culture with MCF-7 cells, the AFP-derived peptides were shown to downregulate cyclin E, SKP2, checkpoint suppressors, cyclin-dependent kinases, and ubiquitin ligases that modulate cyclin E/CdK2 transition from the G1 to the S-phase of the cell cycle. Thus, the experimental data on AFP-CD interaction with cell cycle proteins were consistent with the "in silico" findings.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle , Computational Biology/methods , alpha-Fetoproteins/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Binding Sites/genetics , Cell Cycle Proteins/genetics , Computer Simulation , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Cyclins/metabolism , Humans , MCF-7 Cells , Protein Interaction Domains and Motifs/genetics , Protein Interaction Maps/genetics , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligase Complexes/metabolism , alpha-Fetoproteins/genetics
2.
Tumour Biol ; 36(12): 9857-64, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26162540

ABSTRACT

The concept of a non-secreted cytoplasmic-bound form of alpha-fetoprotein is not a new notion in AFP biological activities. Cytoplasmic AFP (CyAFP) is a long known but forgotten protein in search of a function other than a histochemical biomarker. In this report, CyAFP is presented as an "old" protein with a newly described intracellular function. In 1976, CyAFP was shown to be a product of hepatoma cells utilizing 14Cleucine incorporation and demonstrated by autoradiographic procedures. The synthesis of CyAFP without secretion was demonstrated to occur in both malignant and non-malignant cells encompassing hepatomas, ascite fluid cells, immature rodent uterus, MCF-7 breast cancers, and cytosols from human breast cancer patients. Using computer protein matching and alignments in AFP versus members of the nuclear receptor superfamily, a consecutive series of leucine zipper (heptad) repeats in AFP was previously reported, suggesting the possibility for protein-to-protein interactions. The potential for heptad heterodimerization between protein-binding partners provided the rationale for proposing that CyAFP might have the capability to form molecular hetero-complexes with cytoplasmic based transcription factors. More recent investigations have now provided experimental evidence that CyAFP is capable of colocalizing and interacting with transcription-associated factors. Such proteins can modulate intracellular signaling leading to regulation of transcription factors and initiation of growth in human cancer cells. Although circulating serum AFP is known as a growth-enhancing factor during development, cytoplasmic AFP has a lethal role in the oncogenesis, growth, and metastasis of adult liver cancer.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , DNA-Binding Proteins/genetics , alpha-Fetoproteins/metabolism , Amino Acid Sequence , Breast Neoplasms/pathology , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Female , Humans , MCF-7 Cells , Protein Binding , Signal Transduction , alpha-Fetoproteins/genetics
3.
J Drug Target ; 23(6): 538-51, 2015.
Article in English | MEDLINE | ID: mdl-25766080

ABSTRACT

Recent studies have demonstrated that the carboxyterminal third domain of alpha-fetoprotein (AFP-CD) binds with various ligands and receptors. Reports within the last decade have established that AFP-CD contains a large fragment of amino acids that interact with several different receptor types. Using computer software specifically designed to identify protein-to-protein interaction at amino acid sequence docking sites, the computer searches identified several types of scavenger-associated receptors and their amino acid sequence locations on the AFP-CD polypeptide chain. The scavenger receptors (SRs) identified were CD36, CD163, Stabilin, SSC5D, SRB1 and SREC; the SR-associated receptors included the mannose, low-density lipoprotein receptors, the asialoglycoprotein receptor, and the receptor for advanced glycation endproducts (RAGE). Interestingly, some SR interaction sites were localized on the AFP-derived Growth Inhibitory Peptide (GIP) segment at amino acids #480-500. Following the detection studies, a structural subdomain analysis of both the receptor and the AFP-CD revealed the presence of epidermal growth factor (EGF) repeats, extracellular matrix-like protein regions, amino acid-rich motifs and dimerization subdomains. For the first time, it was reported that EGF-like sequence repeats were identified on each of the three domains of AFP. Thereafter, the localization of receptors on specific cell types were reviewed and their functions were discussed.


Subject(s)
Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Receptors, Scavenger/chemistry , Receptors, Scavenger/metabolism , alpha-Fetoproteins/chemistry , alpha-Fetoproteins/metabolism , Amino Acid Sequence , Binding Sites , Humans , Models, Molecular
4.
Tumour Biol ; 35(8): 7397-402, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24916573

ABSTRACT

The mucin family of proteins is largely expressed on sedentary epithelial cells lining the gastrointestinal, pulmonary, and reproductive tracts and their associated organs and malignant tumors. It is less well-known that mucins are also expressed on circulatory cells of the immune and inflammatory systems, such as monocytes, macrophages, leukemic, and lymphoma cells. The epithelial mucins function in (a) protection and lubrication of mucosal linings, (b) cell adhesion and cell-to-cell contact, (c) cell migration and metastasis, and (d) signal transduction. It would be logical to presume that mucins expressed on circulating mononuclear cells could perform similar functions. Recently, it was proposed that the alpha-fetoprotein (AFP) receptor, known to be present on solid epithelial-derived malignant tumor cells, can be identified as a mucin glycoprotein. Interestingly, it was also reported that AFP binds to a receptor on circulating cells and sedentary tumor cells of lymphoreticular origin, especially monocytes associated with lymphomas and leukemias. The primary objective of the present commentary is to present literature-based evidence that some of the cell surface mucins on sedentary epithelial tumor cells and certain mucins expressed on circulating monocytes/macrophages are identical to the AFP receptor. The secondary objective is to discuss the role of AFP and its derived peptides in the growth suppression of adenocarcinomas and lymphomas using the AFP-mucin receptor concept as a key to the mechanism of tumor growth inhibition.


Subject(s)
Adenocarcinoma/chemistry , Macrophages/chemistry , Monocytes/chemistry , Receptors, Cell Surface/analysis , Receptors, Peptide/analysis , Humans , Mucin-1/physiology , alpha-Fetoproteins/physiology
5.
Tumour Biol ; 34(3): 1317-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23446764

ABSTRACT

The identification of a tumor cell receptor for alpha-fetoprotein (AFP) has long been sought in the field of medicine. The uptake and endocytosis of AFP by rat tumor cells in 1983 sparked a series of confirmatory reports which were extended to include multiple tumor types in rats, mice, and humans. The following year, French investigators characterized the binding properties of the AFP receptor but they did not purify and characterize the receptor. It was not until 1991-1992 that an AFP receptor was partially purified and characterized from both human monocytes and breast cancer cells. By 1993, monoclonal antibodies had been raised against the "AFP receptor" derived from breast cancer extracts with claims that the receptor was a widespread oncoprotein biomarker for cancer. To date, that receptor has yet to be identified due to its complex multimeric structure and carbohydrate composition. The present report will review the literature of the multiple AFP receptors previously including their cellular uptake, transmembrane passage, and partial biochemical characterization. . In addition, evidence derived from computer modeling, proteolytic/fragmentation cleavage patterns, domain structure analysis, and protein binding software analysis will be presented in a proposed identification of a widespread protein/gene family of transmembrane proteins which fits many, if not most, of the criteria attributed to the AFP receptor. The proposed receptor protein family is tentatively identified as an epithelial cell surface mucin constituting one (or more) of many classes of single-pass transmembrane proteins. Present data do not support the concept that the AFP receptor is a "universal" tumor receptor and/or biomarker, but rather a widespread mucin protein that functions primarily in protecting and lubricating epithelial mucosal layers, and engaging in signal transduction; the mucin only binds AFP as a molecule serving in a subordinate or ancillary function.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Mucins/metabolism , Receptors, Cell Surface/metabolism , alpha-Fetoproteins/metabolism , Animals , Humans , Mice , Rats
6.
Curr Mol Med ; 11(7): 564-81, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21707514

ABSTRACT

Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide, with a poor prognosis and limited therapeutic options. Due to its overexpression in the majority of HCCs, alpha-fetoprotein (AFP) represents one of the most useful markers for hepatocarcinomas and for monitoring patients' response to therapy. Although it was earlier reported that AFP has immunosuppressive properties, it has been recently demonstrated that AFP induces spontaneous T and B cells responses in HCC patients. The characterization of AFP-immunogenic epitopes gives the opportunity to design AFP-based cancer vaccines for human HCC. The activity of AFP-based vaccines has been investigated in HCC mouse models in order to develop novel strategies to treat patients with HCC. This review will discuss the rationale for using the AFP-based vaccination strategy and recent results corroborating the usefulness of AFP vaccines as a potential tool for cancer therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Cancer Vaccines/immunology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/immunology , Liver Neoplasms/therapy , alpha-Fetoproteins/immunology , Animals , Cancer Vaccines/therapeutic use , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/metabolism , Mice , alpha-Fetoproteins/metabolism
7.
J Drug Target ; 18(8): 575-88, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20151941

ABSTRACT

The alpha-fetoprotein derived growth inhibitory peptide (GIP) is a 34-amino acid peptide composed of three biologically active subfragments. GIP-34 and its three constituent segments have been synthesized, purified, and studied for biological activity. The GIP-34 and GIP-8 have been characterized as anticancer therapeutic peptides. An multicenter study was initiated to elucidate the means by which these peptide drugs could be targeted to tumor cells. The study first established which cancer types were specifically targeted by the GIP peptides in both in vitro and in vivo investigations. It was next demonstrated that radiolabeled peptide ((125)I GIP-34) is specifically localized to rodent breast tumors at 24 h post-injection. The radionuclide studies also provided evidence for a proposed cell surface receptor; this was confirmed in a further study using fluorescent-labeled GIP-nanobeads which localized at the plasma membrane of MCF-7 breast cancer cells. Finally, it was readily demonstrated that GIP conjugated to either fluorescein or doxorubicin (DOX) underwent tumor cell uptake; subsequently, DOX-GIP conjugates induced cytotoxic cell destruction indicating the utility of GIP segments as cancer therapeutic agents. Following a discussion of the preceding results, a candidate cell surface receptor family was proposed which correlated with previous published reports for a putative AFP/GIP receptor.


Subject(s)
Antineoplastic Agents/administration & dosage , Growth Inhibitors/administration & dosage , Peptide Fragments/administration & dosage , alpha-Fetoproteins/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cell Line, Tumor , Drug Delivery Systems , Growth Inhibitors/chemistry , Growth Inhibitors/metabolism , Humans , Multicenter Studies as Topic , Peptide Fragments/chemistry , Peptide Fragments/metabolism , alpha-Fetoproteins/administration & dosage , alpha-Fetoproteins/metabolism
8.
Gynecol Obstet Invest ; 69(2): 128-30, 2010.
Article in English | MEDLINE | ID: mdl-19996601

ABSTRACT

Fetal alcohol spectrum disorder (FASD) is the leading cause of non-genetic mental retardation in the USA, possibly exceeding even Down syndrome, which is currently approaching 1 in 500 live births. Alcohol consumption during pregnancy results in brain, craniofacial and heart defects, neurotoxicity, and immune dysfunction. The preferred action taken to prevent alcohol consumption during pregnancy is abstinence. However, the detection, diagnosis, and treatment of FASD remain a major public health need in this country and throughout the world. The biochemical molecules involved in the developmental abnormalities encompass a vast array of signal transduction and synaptic pathways which involve neurotransmitters and neurotrophic peptides. Recent advances in medicine-based therapies for FASD have been reported, and include the use of small molecule agonists, antagonists, and competitive inhibitors. Since biomarkers for FASD have previously been identified in clinical research reports, multicenter screening feasibility studies now seem warranted and could be initiated following adequate funding, protocols, procedures, and institutional review board approvals.


Subject(s)
Fetal Alcohol Spectrum Disorders/diagnosis , Prenatal Diagnosis/ethics , Female , Fetal Alcohol Spectrum Disorders/prevention & control , Humans , Infant, Newborn , Pregnancy , Prenatal Exposure Delayed Effects/diagnosis , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/prevention & control
9.
Anticancer Res ; 26(4B): 3071-6, 2006.
Article in English | MEDLINE | ID: mdl-16886636

ABSTRACT

A 34-amino acid peptide has been synthesized based on an amino acid sequence from the third domain of native full-length alpha-fetoprotein, which has been shown to have both antigrowth and anticancer activities. This peptide, known as the growth-inhibitory peptide (GIP), has two cysteine residues and demonstrates reduced antigrowth activity after long-term storage, presumably due to disulfide bond formation. The disulfide bridge problem was addressed by replacing the two naturally occurring cysteines with either glycines, alanines, or serines (to produce the G-, A- and S-peptides, respectively). The non-hydrophobic G- and S-peptides were found to exist as dimers, while the more hydrophobic C- and A-peptides formed trimers in solution under certain conditions of pH and peptide concentration. The A-peptide was already known to display anticancer activity; however, the G- and S-serine analogs have not been studied in depth since they had demonstrated low antigrowth activities in rodent uterine assays. Using both in vivo and in vitro assays, the A-, G- and S-peptides were shown to exhibit various degrees of cancer growth suppression. An in vitro culture assay, using MCF-7 breast cancer cells, demonstrated that both the G- and S-peptides showed modest cancer growth suppression, while the A- analog showed strong inhibition at doses ranging from 10(-5) M to 10(-7) M. In contrast, an in vivo ascites tumor study of all four peptides showed them to have notable activity in the suppression of mouse mammary tumor growth. Overall, our data indicated that physicochemical properties, such as hydrophobicity, oligomeric state and secondary structure, contribute to the anticancer activity of both the active C- peptide and its analogs. In addition, the antigrowth rodent uterine assay was not always predictive of the anticancer potential of the peptide forms, suggesting a difference between the mechanism of peptide action in the antigrowth models and that in the anticancer assay. Notably, the antigrowth assay failed to predict the marked anticancer activity of the analogs against a mammary tumor, indicating that the growth bioassay cannot always be relied upon as a screening protocol.


Subject(s)
Antineoplastic Agents/pharmacology , Growth Inhibitors/pharmacology , Membrane Proteins/chemistry , Membrane Proteins/pharmacology , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatography, High Pressure Liquid , Female , Growth Inhibitors/chemistry , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Uterus/drug effects , Uterus/growth & development
10.
Chemotherapy ; 52(2): 83-90, 2006.
Article in English | MEDLINE | ID: mdl-16498241

ABSTRACT

The present update describes the biological activities of an alpha fetoprotein (AFP)-derived peptide termed the growth-inhibitory peptide (GIP), which is a synthetic 34-amino acid segment produced from the native molecule. The GIP has been shown to be growth-suppressive in both fetal and tumor cells but not in adult cells. Even though its mechanism of action has not been completely elucidated, GIP has been shown to engage in cellular events such as endocytosis, cellular aggregation, hemagglution, and cytoskeleton-induced cell shape changes. The GIP was shown to be growth-suppressive in nine different human tumor types and to suppress the spread of tumor infiltrates and metastases in human and mouse mammary cancers. It was further found that the oligomeric state (cyclic vs. linear configuration) of the GIP determined its biological and anticancer efficacy. The combined properties of tumor growth suppression and reduction of tumor metastases represent promising areas for GIP as a chemotherapeutic agent. It was concluded that the GIP derived from full-length AFP represents a growth-inhibitory motif possessing intrinsic properties that allow it to interact in cell surface events such as tumor proliferation, progression, and metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Mammary Neoplasms, Experimental/pathology , Peptide Fragments/pharmacology , alpha-Fetoproteins/chemistry , alpha-Fetoproteins/pharmacology , Animals , Ascites/drug therapy , Ascites/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Nude , Neoplasm Metastasis , Peptide Fragments/chemistry , Quantitative Structure-Activity Relationship , Transplantation, Heterologous
11.
Cancer Metastasis Rev ; 24(3): 441-67, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16258731

ABSTRACT

This review surveys the biological activities of an alpha-fetoprotein (AFP) derived peptide termed the Growth Inhibitory Peptide (GIP), which is a synthetic 34 amino acid segment produced from the full length 590 amino acid AFP molecule. The GIP has been shown to be growth-suppressive in both fetal and tumor cells but not in adult terminally-differentiated cells. The mechanism of action of this peptide has not been fully elucidated; however, GIP is highly interactive at the plasma membrane surface in cellular events such as endocytosis, cell contact inhibition and cytoskeleton-induced cell shape changes. The GIP was shown to be growth-suppressive in nine human tumor types and to suppress the spread of tumor infiltrates and metastases in human and mouse mammary cancers. The AFP-derived peptide and its subfragments were also shown to inhibit tumor cell adhesion to extracellular matrix (ECM) proteins and to block platelet aggregation; thus it was expected that the GIP would inhibit cell spreading/migration and metastatic infiltration into host tissues such as lung and pancreas. It was further found that the cyclic versus linear configuration of GIP determined its biological and anti-cancer efficacy. Genbank amino acid sequence identities with a variety of integrin alpha/beta chain proteins supported the GIP's linkage to inhibition of tumor cell adhesion and platelet aggregation. The combined properties of tumor growth suppression, prevention of tumor cell-to-ECM adhesion, and inhibition of platelet aggregation indicate that tumor-to-platelet interactions present promising targets for GIP as an anti-metastatic agent. Finally, based on cholinergic studies, it was proposed that GIP could influence the enzymatic activity of membrane acetylcholinesterases during tumor growth and metastasis. It was concluded that the GIP derived from full-length AFP represents a growth inhibitory motif possessing instrinsic properties that allow it to interfere in cell surface events such as adhesion, migration, metastasis, and aggregation of tumor cells.


Subject(s)
Biological Products/therapeutic use , Growth Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/therapy , Peptides/therapeutic use , alpha-Fetoproteins/therapeutic use , Amino Acid Sequence , Animals , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Cytoskeleton/metabolism , Disease Progression , Endocytosis , Growth Inhibitors/chemistry , Humans , Mice , Models, Biological , Molecular Sequence Data , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Peptides/chemistry , Platelet Aggregation , Protein Conformation , Protein Structure, Tertiary , Time Factors
12.
Fetal Diagn Ther ; 18(5): 360-9, 2003.
Article in English | MEDLINE | ID: mdl-12913349

ABSTRACT

Both insulin and estrogen are well recognized as growth-promoting substances at physiological concentrations, but they function as teratogens at high doses. Both agents can affect alterations in fetal and maternal serum human alpha-fetoprotein (HAFP) levels during pregnancy. In the present study, we have employed animal models of both insulin and estrogen fetotoxicity and teratogenicity in order to study the growth-regulatory properties of HAFP and its derived peptides (HAFP/PEP). We report here the effects of HAFP/PEP on fetotoxicity, congenital malformations, and growth retardation in developing chick and murine fetuses. In the insulin model, HAFP/PEP were effective in reducing both fetal mortality and anatomic anomalies, with the result that growth-retarded fetuses were produced. With HAFP/PEP treatment, fetal demise was reduced by as much as 73 and 63% in murine and chick fetuses, respectively, while fetal anomalies were diminished by 50% during chick development. Genebank searches of identity/similarity in a HAFP/PEP fragment identified matches with a number of proteins associated with glucose, pH, ionic, osmotic, and oxidative stresses, and with heat shock, in addition to stress proteins related to protein folding/unfolding processes. It was proposed that the peptide segment on HAFP may represent a topographic 'hotspot', sensitive to stress/shock conditions, which exhibits a propensity for conformational alteration in the tertiary structure of the fetal protein.


Subject(s)
Estrogens/toxicity , Fetal Growth Retardation/physiopathology , Hypoglycemic Agents/toxicity , Insulin/toxicity , Teratogens/toxicity , alpha-Fetoproteins/pharmacology , Albumins/genetics , Amino Acid Sequence , Animals , Chick Embryo , Chickens , Female , Fetal Growth Retardation/chemically induced , Fetal Growth Retardation/drug therapy , Heat-Shock Proteins/genetics , Mice , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Pregnancy , alpha-Fetoproteins/genetics
13.
J Pept Res ; 61(4): 213-8, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12605606

ABSTRACT

A chemically synthesized 34-amino acid peptide, an analog, and a fragment of the peptide have been purified and studied. Biophysical studies were carried out to determine some of the metal ion binding properties of the original peptide and an analog of this parent peptide, in which the two histidine residues were replaced by alanines. As shown by visible absorption spectroscopy, Co (II) forms a complex with the parent peptide, but not with the analog peptide, and one or two histidines in the parent peptide are ligands for Co (II) ion binding. The effects on disulfide bond formation in the peptide by Zn (II) and Co (II) ions were also examined for this analog. Anti-growth assays were performed using the original cysteine-containing peptide with Zn (II) ion complexed to the peptide through the two cysteine residues. These rat uterine growth assays showed that the complexing of Zn (II) ion to the peptide maintained the anti-growth activity of the peptide, while gel-filtration experiments showed the zinc ions maintained the peptide in its anti-growth form indefinitely in solution. A saliently important part of this research was the discovery that a fragment of the peptide consisting of a middle sequence of 14 amino acids was found to have significant anti-growth activity in the rat uterine assay. Its activity suggested that this fragment might be considered a viable candidate for testing in anti-cancer protocols.


Subject(s)
Peptide Fragments/pharmacology , alpha-Fetoproteins/chemistry , Amino Acid Sequence , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Chromatography, Gel , Circular Dichroism , Ligands , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Rats , Zinc/metabolism
14.
Med Hypotheses ; 58(6): 453-61, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12323110

ABSTRACT

Alpha-fetoprotein (AFP) serum levels in man have long been utilized as a tumor marker and as a birth defect screening agent in the clinical laboratory. Although the physiological role of AFP has remained obscure, the stereotypic carrier/transport function of a fetal counterpart to albumin has been attributed to this oncofetal protein. However, reports from a multitude of investigators in the last decade have provided a rationale to reconsider and extend the biological role of AFP to include cell growth modulation during development. Previously, a leucine zipper-like (heptad) motif, which mimicked that found in the steroid/thyroid receptor superfamily, was postulated for portions of the third domain of AFP. The present report proposes the presence of additional potential heterodimerization sites for the nuclear receptor superfamily members and other growth factors in the second and third domains of human AFP.


Subject(s)
Growth Substances/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , alpha-Fetoproteins/metabolism , Amino Acid Sequence , Animals , Dimerization , Humans , Molecular Sequence Data , Protein Binding , Sequence Homology, Amino Acid , alpha-Fetoproteins/chemistry
15.
Pediatrics ; 108(6): 1370-3, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11731663

ABSTRACT

Docosahexaenoic acid, found lacking in animal models of cystic fibrosis, has been proposed as a dietary supplement therapy for this genetic disorder. Alpha-fetoprotein, which binds and transports docosahexaenoic acid, may be a useful marker to improve the management and follow-up in newborn screening programs for cystic fibrosis, because only 20% of such infants are diagnosed at birth.


Subject(s)
Cystic Fibrosis/prevention & control , Docosahexaenoic Acids/metabolism , alpha-Fetoproteins/metabolism , Biomarkers/blood , Cystic Fibrosis/blood , Cystic Fibrosis/metabolism , Dietary Supplements , Docosahexaenoic Acids/therapeutic use , Humans , Infant , Infant, Newborn , Neonatal Screening
16.
Biochim Biophys Acta ; 1528(2-3): 127-34, 2001 Oct 03.
Article in English | MEDLINE | ID: mdl-11687299

ABSTRACT

A 34-amino-acid peptide has been chemically synthesized based on a sequence from human alpha-fetoprotein. The purified peptide is active in anti-growth assays when freshly prepared in pH 7.4 buffer at 0.20 g/l, but this peptide slowly becomes inactive. This functional change is proven by mass spectrometry to be triggered by the formation of an intrapeptide disulfide bond between the two cysteine residues on the peptide. Interpeptide cross-linking does not occur. The active and inactive forms of the peptide have almost identical secondary structures as shown by circular dichroism (CD). Zinc ions bind to the active peptide and completely prevents formation of the inactive form. Cobalt(II) ions also bind to the peptide, and the UV-Vis absorption spectrum of the cobalt-peptide complex shows that: (1) a near-UV sulfur-to-metal-ion charge-transfer band had a molar extinction coefficient consistent with two thiolate bonds to Co(II); (2) the lowest-energy visible d-d transition maximum at 659 nm, also, demonstrated that the two cysteine residues are ligands for the metal ion; (3) the d-d molar extinction coefficient showed that the metal ion-ligand complex was in a distorted tetrahedral symmetry. The peptide has two cysteines, and it is speculated that the other two metal ion ligands might be the two histidines. The Zn(II)- and Co(II)-peptide complexes had similar peptide conformations as indicated by their ultraviolet CD spectra, which differed very slightly from that of the free peptide. Surprisingly, the cobalt ions acted in the reverse of the zinc ions in that, instead of stabilizing anti-growth form of the peptide, they catalyzed its loss. Metal ion control of peptide function is a saliently interesting concept. Calcium ions, in the conditions studied, apparently do not bind to the peptide. Trifluoroethanol and temperature (60 degrees C) affected the secondary structure of the peptide, and the peptide was found capable of assuming various conformations in solution. This conformational flexibility may possibly be related to the biological activity of the peptide.


Subject(s)
Peptides/chemical synthesis , alpha-Fetoproteins/chemistry , Cations , Chromatography, Gel , Circular Dichroism , Cobalt/chemistry , Disulfides/chemistry , Metals/chemistry , Peptides/chemistry , Peptides/physiology , Protein Conformation , Protein Structure, Secondary , Solutions , Spectrophotometry , Spectrophotometry, Ultraviolet , Zinc/chemistry
17.
J Pept Res ; 57(6): 539-46, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11437957

ABSTRACT

A 34-amino acid portion of the third domain of alpha-fetoprotein possesses antigrowth and anticancer activities. Three analogs of this sequence were chemically synthesized, in which the two cysteines of the original sequence were replaced by alanines, glycines or serines. The original cysteine and alanine peptides formed trimers at 0.20 g/L in pH 7.4 phosphate buffer, and the glycine and serine peptides formed dimers. Trimer preparations were more potent in inhibiting estrogen-induced growth in the mouse uterine assays than the two dimeric oligomers. Of salient importance is that the alanine peptide retained its trimeric form in solution much longer than the cysteine peptide. Antigrowth assays were performed starting with stock solutions at a peptide concentration of 0.20 g/L, because at very high peptide concentration (8.0 g/L) the peptides aggregated extensively. All the peptides, although differing in biological activity, had almost identical secondary structures. Unlike alpha-fetoprotein, the three peptides have low amounts of alpha-helix. Trifluoroethanol has the ability to convert peptides into a helical conformation when they have a propensity for that structure. At trifluoroethanol concentrations of 20% and higher, the alanine and glycine peptides were changed into highly helical structures.


Subject(s)
Antineoplastic Agents/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , alpha-Fetoproteins/chemistry , alpha-Fetoproteins/pharmacology , Alanine/analogs & derivatives , Alanine/chemistry , Animals , Chromatography, Gel , Cysteine/analogs & derivatives , Cysteine/chemistry , Dose-Response Relationship, Drug , Epitopes , Estrogens/metabolism , Female , Glycine/analogs & derivatives , Glycine/chemistry , Humans , Hydrogen-Ion Concentration , Mice , Protein Structure, Secondary , Serine/analogs & derivatives , Serine/chemistry , Time Factors , Uterus/drug effects
18.
Exp Biol Med (Maywood) ; 226(5): 377-408, 2001 May.
Article in English | MEDLINE | ID: mdl-11393167

ABSTRACT

Mammalian alpha-fetoprotein (AFP) is classified as a member of the albuminoid gene superfamily consisting of albumin, AFP, vitamin D (Gc) protein, and alpha-albumin. Molecular variants of AFP have long been reported in the biomedical literature. Early studies identified isoelectric pH isoforms and lectin-binding variants of AFP, which differed in their physicochemical properties, but not in amino acid composition. Genetic variants of AFP, differing in mRNA kilobase length, were later extensively described in rodent models during fetal/perinatal stages, carcinogenesis, and organ regeneration. With the advent of monoclonal antibodies in the early 1980s, multiple antigenic epitopes on native AFP were detected and categorized, culminating in the identification of six to seven major epitopes. During this period, various AFP-binding proteins and receptors were reported to inhibit certain AFP immunoreactions. Concomittantly, human and rodent AFP were cloned and the amino acid sequences of the translated proteins were divulged. Once the amino acid composition of the AFP molecule was known, enzymatic fragments could be identified and synthetic peptide segments synthesized. Following discovery of the molten globule form in 1981, the existence of transitory, intermediate forms of AFP were acknowledged and their physiological significance was realized. In the present review, the various isoforms and variants of AFP are discussed in light of their potential biological relevance.


Subject(s)
alpha-Fetoproteins/chemistry , alpha-Fetoproteins/physiology , Amino Acid Sequence , Animals , Antigenic Variation , Carbohydrates/chemistry , Cell Adhesion , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Epitope Mapping , Epitopes/chemistry , Female , Gene Expression Regulation , Genetic Diseases, Inborn/genetics , Genetic Variation , Growth Substances/physiology , Humans , Hydrogen-Ion Concentration , Immune Tolerance , Maternal-Fetal Exchange , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/physiology , Molecular Sequence Data , Multigene Family , Neoplasms/drug therapy , Neoplasms/prevention & control , Peptide Fragments/chemistry , Pregnancy , Protein Conformation , Protein Isoforms/chemistry , Sequence Homology, Amino Acid , alpha-Fetoproteins/genetics
19.
J Pept Res ; 57(1): 29-38, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11168886

ABSTRACT

A 34-amino acid synthetic peptide was derived from the third domain of human alpha-fetoprotein, and the peptide was shown to inhibit estrogen-stimulated growth. Under certain conditions, however, the peptide lost growth-inhibitory activity. A biophysical study of the peptide was undertaken with a goal of obtaining completely reliable preparations. The peptide was studied using gel-filtration column chromatography as a function of peptide concentration and age of solution, and was found to exhibit complex aggregation behaviors. During the early period (0-3 h) after dissolving lyophilized peptide into pH 7.4 buffer, solutions were composed mostly of trimers. At higher peptide concentrations (> or = 3.0 g/L), the trimers aggregated extensively to a large aggregate (minimum size approximately 102 peptides). At 5.0-8.0 g/L, these large aggregates increased in size (up to approximately 146 peptides) until trimers were largely exhausted from solution. During the later times (>3 h) after sample preparation, the trimeric oligomer of the peptide dissociated slowly to form dimers for samples at 0.10-3.0 g/L. After their build-up, a very small number of dimers associated to form hexamers. Disulfide bonds stabilized the dimers as indicated by the conversion of dimers to trimers upon the addition of a reducing agent, and the failure of dimers to form in the presence of reducing agent. Reducing agent did not affect trimer or large aggregate formation. Trimers were found to be active in an assay monitoring inhibition of estrogen-stimulated growth, whereas dimers and large aggregates were inactive. The two cysteines in the peptide were modified to either S-methylcysteine or S-(2-aminoethyl)cysteine, and both derivatives showed significant growth-inhibition activity. A serine analog in which both cysteines were replaced had very different aggregation behavior than the cysteine peptide and lacked its growth inhibitory ability. Peptide aggregation is critically important in establishing the ability of the peptide to inhibit growth and have anticancer activity, but the state of its two cysteines is of little influence.


Subject(s)
Antineoplastic Agents/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , alpha-Fetoproteins/chemistry , alpha-Fetoproteins/pharmacology , Acetylcysteine/analogs & derivatives , Acetylcysteine/chemistry , Animals , Chromatography, Gel , Cysteine/analogs & derivatives , Cysteine/chemistry , Disulfides , Dose-Response Relationship, Drug , Epitopes , Estrogens/metabolism , Female , Humans , Hydrogen-Ion Concentration , Mice , Peptide Biosynthesis , Protein Structure, Tertiary , Time Factors , Uterus/drug effects
20.
Expert Opin Investig Drugs ; 10(6): 1063-73, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11772235

ABSTRACT

Peptides act as effector agents that regulate and/or mediate physiological processes, serving as hormones, neurotransmitters and signal transducing factors. The low molecular weight peptides affect receptor-mediated events, which influence cardiovascular, gastrointestinal and neurocranial systems. While some peptides have been marketed as drugs, many have served as leads or templates for the development of non-peptide drugs that mimic peptide actions. This review presents the advantages and disadvantages of using peptides as drugs that bind as ligands to cell-surface receptors and considers their applications in such events. The value of both the peptides and their mimics is based on their participation in the biomodulation of physiological processes, which frequently employ scaffolding proteins acting in a cascading sequence of protein-to-protein interactions. The peptides bind to G-coupled surface receptors to initiate a signal that is transduced to the interior of the cell through multiple layers of phosphorylating enzymes and binding proteins. Peptides have been further employed to identify the molecular targets of signal transduction, the uncoupling of which might provide a means for various disease therapies. The exploitation of such peptide-mediated signal pathways, which are of primary importance to tumour cells, may provide an attractive strategy for anticancer therapy in the future.


Subject(s)
GTP-Binding Proteins/metabolism , Ligands , Peptides/metabolism , Peptides/therapeutic use , Receptors, Peptide/metabolism , Signal Transduction/drug effects , Animals , Humans , Peptides/pharmacology , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...