1
|
El-Fakharany EM, El-Gendi H, Abdel-Wahhab K, Abu-Serie MM, El-Sahra DG, Ashry M. Therapeutic efficacy of α-lactalbumin coated oleic acid based liposomes against colorectal carcinoma in Caco-2 cells and DMH-treated albino rats. J Biomol Struct Dyn 2024; 42:9220-9234. [PMID: 37624964 DOI: 10.1080/07391102.2023.2250452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
Colorectal cancer (CRC) is a malignant tumor recognized as a major cause of morbidity and mortality throughout the world. Therefore, novel liposomes of oleic acid coated with camel α-lactalbumin (α-LA coated liposomes) were developed for their potential antitumor activity against CRC, both in vitro and in DMH-induced CRC-modeled animal. In vitro results indicated the high safety of α-LA coated liposomes towards normal human cells with potent antitumor activity against Caco-2 cells at an IC50 value of 57.01 ± 3.55 µM with selectivity index of 6.92 ± 0.48. This antitumor activity has been attributed to induction of the apoptotic mechanism, as demonstrated by nuclear condensation and arrest of Caco-2 cells in sub-G1 populations. α-LA coated liposomes also revealed a significant up-regulation of the p53 gene combined with a down-regulation of the Bcl2 gene. Moreover, in vivo results revealed that treatment of induced-CRC modeled animals with α-LA coated liposomes for six weeks markedly improved the CRC-disorders; this was achieved from the significant reduction in the values of AFP, CEA, CA19.9, TNF-α, IL-1β, MDA, and NO coupled with remarkable rise in SOD, GPx, GSH, CAT, and CD4+ levels. The histopathological findings asserted the therapeutic potential of α-LA coated liposomes in the treatment of CRC. Therefore, the present results proved the antitumor activity of α-LA coated liposomes against CRC through the restoration of impaired oxidative stress, improved immune response, and reduced inflammation.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Esmail M El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA- City), Alexandria, Egypt
| | - Hamada El-Gendi
- Bioprocess Development Department, GEBRI, SRTA-City, Alexandria, Egypt
| | | | - Marwa M Abu-Serie
- Medical Biotechnology Department, GEBRI, SRTA-City, Alexandria, Egypt
| | - Doaa Galal El-Sahra
- Medical Surgical Nursing Department, Faculty of Nursing, Modern University for Technology and Information, Cairo, Egypt
| | - Mahmoud Ashry
- Zoology Department, Faculty of Science, Al-Azhar University, Assuit, Egypt
| |
Collapse
|
2
|
Imamura H, Ooishi A, Honda S. Getting Smaller by Denaturation: Acid-Induced Compaction of Antibodies. J Phys Chem Lett 2023; 14:3898-3906. [PMID: 37093025 PMCID: PMC10150727 DOI: 10.1021/acs.jpclett.3c00258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/07/2023] [Indexed: 05/03/2023]
Abstract
Protein denaturation is a ubiquitous process that occurs both in vitro and in vivo. While our molecular understanding of the denatured structures of proteins is limited, it is commonly accepted that the loss of unique intramolecular contacts makes proteins larger. Herein, we report compaction of the immunoglobulin G1 (IgG1) protein upon acid denaturation. Small-angle X-ray scattering coupled with size exclusion chromatography revealed that IgG1 radii of gyration at pH 2 were ∼75% of those at a neutral pH. Scattering profiles showed a compact globular shape, supported by analytical ultracentrifugation. The acid denaturation of proteins with a decrease in size is energetically costly, and acid-induced compaction requires an attractive force for domain reorientation. Such intramolecular aggregation may be widespread in immunoglobulin proteins as noncanonical structures. Herein, we discuss the potential biological significance of these noncanonical structures of antibodies.
Collapse
Affiliation(s)
- Hiroshi Imamura
- Biomedical
Research Institute, National Institute of
Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
- Department
of Applied Chemistry, College of Life Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga 525-8577, Japan
- Department
of Bio-Science, Nagahama Institute of Bio-Science
and Technology, 1266 Tamura, Nagahama, Shiga 526-0829, Japan
| | - Ayako Ooishi
- Biomedical
Research Institute, National Institute of
Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Shinya Honda
- Biomedical
Research Institute, National Institute of
Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| |
Collapse
|
3
|
Proteins and their functionalization for finding therapeutic avenues in cancer: Current status and future prospective. Biochim Biophys Acta Rev Cancer 2023; 1878:188862. [PMID: 36791920 DOI: 10.1016/j.bbcan.2023.188862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 02/15/2023]
Abstract
Despite the remarkable advancement in the health care sector, cancer remains the second most fatal disease globally. The existing conventional cancer treatments primarily include chemotherapy, which has been associated with little to severe side effects, and radiotherapy, which is usually expensive. To overcome these problems, target-specific nanocarriers have been explored for delivering chemo drugs. However, recent reports on using a few proteins having anticancer activity and further use of them as drug carriers have generated tremendous attention for furthering the research towards cancer therapy. Biomolecules, especially proteins, have emerged as suitable alternatives in cancer treatment due to multiple favourable properties including biocompatibility, biodegradability, and structural flexibility for easy surface functionalization. Several in vitro and in vivo studies have reported that various proteins derived from animal, plant, and bacterial species, demonstrated strong cytotoxic and antiproliferative properties against malignant cells in native and their different structural conformations. Moreover, surface tunable properties of these proteins help to bind a range of anticancer drugs and target ligands, thus making them efficient delivery agents in cancer therapy. Here, we discuss various proteins obtained from common exogenous sources and how they transform into effective anticancer agents. We also comprehensively discuss the tumor-killing mechanisms of different dietary proteins such as bovine α-lactalbumin, hen egg-white lysozyme, and their conjugates. We also articulate how protein nanostructures can be used as carriers for delivering cancer drugs and theranostics, and strategies to be adopted for improving their in vivo delivery and targeting. We further discuss the FDA-approved protein-based anticancer formulations along with those in different phases of clinical trials.
Collapse
|
4
|
Carboxyl Group-Modified Myoglobin Induces TNF-α-Mediated Apoptosis in Leukemia Cells. Pharmaceuticals (Basel) 2022; 15:ph15091066. [PMID: 36145287 PMCID: PMC9501283 DOI: 10.3390/ph15091066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
Previous studies have shown that chemical modification may increase the activity of proteins or confer novel activity to proteins. Some studies have indicated that myoglobin (Mb) is cytotoxic; however, the underlying mechanisms remain unclear. In this study, we investigated whether chemical modification of the carboxyl group by semicarbazide could promote the Mb cytotoxicity in human leukemia U937 cells and the underlying mechanism of semicarbazide-modified myoglobin (SEM-Mb)-induced U937 cell death. The semicarbazide-modified Mb (SEM-Mb) induced U937 cell apoptosis via the production of cleaved caspase-8 and t-Bid, while silencing of FADD abolished this effect. These findings suggest that SEM-Mb can induce U937 cell death by activating the death receptor-mediated pathway. The SEM-Mb inhibited miR-99a expression, leading to increased NOX4 mRNA and protein expression, which promoted SIRT3 degradation, and, in turn, induced ROS-mediated p38 MAPK phosphorylation. Activated p38 MAPK stimulated miR-29a-dependent tristetraprolin (TTP) mRNA decay. Downregulation of TTP slowed TNF-α mRNA turnover, thereby increasing TNF-α protein expression. The SEM-Mb-induced decrease in cell viability and TNF-α upregulation were alleviated by abrogating the NOX4/SIRT3/ROS/p38 MAPK axis or ectopic expression of TTP. Taken together, our results demonstrated that the NOX4/SIRT3/p38 MAPK/TTP axis induces TNF-α-mediated apoptosis in U937 cells following SEM-Mb treatment. A pathway regulating p38 MAPK-mediated TNF-α expression also explains the cytotoxicity of SEM-Mb in the human leukemia cell lines HL-60, THP-1, K562, Jurkat, and ABT-199-resistant U937. Furthermore, these findings suggest that the carboxyl group-modified Mb is a potential structural template for the generation of tumoricidal proteins.
Collapse
|
5
|
Tashiro D, Suetaka S, Sato N, Ooka K, Kunihara T, Kudo H, Inatomi J, Hayashi Y, Arai M. Intron-Encoded Domain of Herstatin, An Autoinhibitor of Human Epidermal Growth Factor Receptors, Is Intrinsically Disordered. Front Mol Biosci 2022; 9:862910. [PMID: 35573740 PMCID: PMC9100580 DOI: 10.3389/fmolb.2022.862910] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/31/2022] [Indexed: 11/17/2022] Open
Abstract
Human epidermal growth factor receptors (HER/ERBB) form dimers that promote cell proliferation, migration, and differentiation, but overexpression of HER proteins results in cancer. Consequently, inhibitors of HER dimerization may function as effective antitumor drugs. An alternatively spliced variant of HER2, called herstatin, is an autoinhibitor of HER proteins, and the intron 8-encoded 79-residue domain of herstatin, called Int8, binds HER family receptors even in isolation. However, the structure of Int8 remains poorly understood. Here, we revealed by circular dichroism, NMR, small-angle X-ray scattering, and structure prediction that isolated Int8 is largely disordered but has a residual helical structure. The radius of gyration of Int8 was almost the same as that of fully unfolded states, although the conformational ensemble of Int8 was less flexible than random coils. These results demonstrate that Int8 is intrinsically disordered. Thus, Int8 is an interesting example of an intrinsically disordered region with tumor-suppressive activity encoded by an intron. Furthermore, we show that the R371I mutant of Int8, which is defective in binding to HER2, is prone to aggregation, providing a rationale for the loss of function.
Collapse
Affiliation(s)
- Daisuke Tashiro
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Shunji Suetaka
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Nao Sato
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Koji Ooka
- Department of Physics, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Tomoko Kunihara
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Hisashi Kudo
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Junichi Inatomi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuuki Hayashi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Munehito Arai
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
- Department of Physics, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- *Correspondence: Munehito Arai,
| |
Collapse
|
6
|
Dietary Linolenic Acid Increases Sensitizing and Eliciting Capacities of Cow's Milk Whey Proteins in BALB/c Mice. Nutrients 2022; 14:nu14040822. [PMID: 35215473 PMCID: PMC8877816 DOI: 10.3390/nu14040822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 02/01/2023] Open
Abstract
α-Lactalbumin (BLA) and β-lactoglobulin (BLG) are the major whey proteins causing allergic reactions. Polyunsaturated fatty acids (PUFAs) stand among the extrinsic factors of the food matrix that can bind BLA and BLG and change their bioactivities, but their contribution to change the allergenic properties of these proteins has not been investigated. Here, we aimed to determine how PUFAs influence BLA and BLG to sensitize and trigger allergic responses in BALB/c mice. First, tricine-SDS-PAGE and spectroscopic assays identified that α-linolenic acid (ALA, as a proof-of-concept model) can induce BLA and BLG to form cross-linked complexes and substantially modify their conformation. Then, BALB/c mice (n = 10/group) were orally sensitized and challenged with BLA and BLG or ALA-interacted BLA and BLG, respectively. Allergic reactions upon oral challenge were determined by measuring clinical allergic signs, specific antibodies, levels of type-1/2 cytokines, the status of mast cell activation, and percentage of cell populations (B and T cells) in different tissues (PP, MLN, and spleen). Overall, systemic allergic reaction was promoted in mice gavage with ALA-interacted BLA and BLG by disrupting the Th1/Th2 balance toward a Th2 immune response with the decreased number of Tregs. Enhanced induction of Th2-related cytokines, as well as serum-specific antibodies and mast cell activation, was also observed. In this study, we validated that ALA in the food matrix promoted both the sensitization and elicitation of allergic reactions in BALB/c mice.
Collapse
|
7
|
Chetta KE, Alcorn JL, Baatz JE, Wagner CL. Cytotoxic Lactalbumin-Oleic Acid Complexes in the Human Milk Diet of Preterm Infants. Nutrients 2021; 13:4336. [PMID: 34959888 PMCID: PMC8707396 DOI: 10.3390/nu13124336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/23/2021] [Accepted: 11/28/2021] [Indexed: 12/13/2022] Open
Abstract
Frozen storage is necessary to preserve expressed human milk for critically ill and very preterm infants. Milk pasteurization is essential for donor milk given to this special population. Due to these storage and processing conditions, subtle changes occur in milk nutrients. These changes may have clinical implications. Potentially, bioactive complexes of unknown significance could be found in human milk given to preterm infants. One such complex, a cytotoxic α-lactalbumin-oleic acid complex named "HAMLET," (Human Alpha-Lactalbumin Made Lethal to Tumor cells) is a folding variant of alpha-lactalbumin that is bound to oleic acid. This complex, isolated from human milk casein, has specific toxicity to both carcinogenic cell lines and immature non-transformed cells. Both HAMLET and free oleic acid trigger similar apoptotic mechanisms in tissue and stimulate inflammation via the NF-κB and MAPK p38 signaling pathways. This protein-lipid complex could potentially trigger various inflammatory pathways with unknown consequences, especially in immature intestinal tissues. The very preterm population is dependent on human milk as a medicinal and broadly bioactive nutriment. Therefore, HAMLET's possible presence and bioactive role in milk should be addressed in neonatal research. Through a pediatric lens, HAMLET's discovery, formation and bioactive benefits will be reviewed.
Collapse
Affiliation(s)
- Katherine E. Chetta
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Medical University of South Carolina, Shawn Jenkins Children’s Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA; (J.E.B.); (C.L.W.)
| | - Joseph L. Alcorn
- Department of Pediatrics, Division of Neonatology and Pediatric Research Center, The University of Texas Health & Science Center at Houston, 6631 Fannin Street MSB 3.252, Houston, TX 77030, USA;
| | - John E. Baatz
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Medical University of South Carolina, Shawn Jenkins Children’s Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA; (J.E.B.); (C.L.W.)
| | - Carol L. Wagner
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Medical University of South Carolina, Shawn Jenkins Children’s Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, USA; (J.E.B.); (C.L.W.)
| |
Collapse
|
8
|
Meng X, Zeng Z, Gao J, Tong P, Wu Y, Li X, Chen H. Conformational changes in bovine α-lactalbumin and β-lactoglobulin evoked by interaction with C18 unsaturated fatty acids provide insights into increased allergic potential. Food Funct 2020; 11:9240-9251. [PMID: 33034612 DOI: 10.1039/d0fo02028a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Bovine α-lactalbumin (BLA) and β-lactoglobulin (BLG) are the most common and severe food allergens in milk and they can bind C18 unsaturated fatty acids (UFAs) and their bioactivities were changed. This study aims to determine the effects of C18 UFAs on the structures of BLA and BLG and their allergic properties, such as antigenicity and allergenicity. We reveal that C18 UFAs can efficiently promote the gradual unfolding of the structures of BLA and BLG and increase their hydrophobicity. Moreover, the IgG binding ability and the expression of IgG-dependent activation marker CD200R3 on basophils were remarkably promoted after C18 UFA treatment. Finally, we also observed that C18 UFAs can enhance the IgE binding ability and the degranulation capacity of human basophil KU812 cells (intracellular Ca2+, histamine, β-Hex, and IL-6). Collectively, these results suggested that C18 UFAs changed the structures of BLA and BLG, which contributed to their increased allergic potential.
Collapse
Affiliation(s)
- Xuanyi Meng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China. and Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China
| | - Zheling Zeng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China. and Jiangxi Province Key Laboratory of Edible and Medicinal Resources Exploitation, Nanchang University, Nanchang 330031, China
| | - Jinyan Gao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China. and School of Food Science & Technology, Nanchang University, Nanchang 330031, China
| | - Ping Tong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China.
| | - Yong Wu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China. and Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China
| | - Xin Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China. and School of Food Science & Technology, Nanchang University, Nanchang 330031, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China. and Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China
| |
Collapse
|
9
|
α-Lactalbumin, Amazing Calcium-Binding Protein. Biomolecules 2020; 10:biom10091210. [PMID: 32825311 PMCID: PMC7565966 DOI: 10.3390/biom10091210] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023] Open
Abstract
α-Lactalbumin (α-LA) is a small (Mr 14,200), acidic (pI 4–5), Ca2+-binding protein. α-LA is a regulatory component of lactose synthase enzyme system functioning in the lactating mammary gland. The protein possesses a single strong Ca2+-binding site, which can also bind Mg2+, Mn2+, Na+, K+, and some other metal cations. It contains several distinct Zn2+-binding sites. Physical properties of α-LA strongly depend on the occupation of its metal binding sites by metal ions. In the absence of bound metal ions, α-LA is in the molten globule-like state. The binding of metal ions, and especially of Ca2+, increases stability of α-LA against the action of heat, various denaturing agents and proteases, while the binding of Zn2+ to the Ca2+-loaded protein decreases its stability and causes its aggregation. At pH 2, the protein is in the classical molten globule state. α-LA can associate with membranes at neutral or slightly acidic pH at physiological temperatures. Depending on external conditions, α-LA can form amyloid fibrils, amorphous aggregates, nanoparticles, and nanotubes. Some of these aggregated states of α-LA can be used in practical applications such as drug delivery to tissues and organs. α-LA and some of its fragments possess bactericidal and antiviral activities. Complexes of partially unfolded α-LA with oleic acid are cytotoxic to various tumor and bacterial cells. α-LA in the cytotoxic complexes plays a role of a delivery carrier of cytotoxic fatty acid molecules into tumor and bacterial cells across the cell membrane. Perhaps in the future the complexes of α-LA with oleic acid will be used for development of new anti-cancer drugs.
Collapse
|
10
|
Pedersen JN, Frislev HKS, Pedersen JS, Otzen D. Structures and mechanisms of formation of liprotides. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140505. [PMID: 32721568 DOI: 10.1016/j.bbapap.2020.140505] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022]
Abstract
Many proteins form complexes called liprotides with oleic acid and other cis-fatty acids under conditions where the protein is partially unfolded. The complexes vary in structure depending on the ratio of protein and lipid, but the most common structural organization is the core-shell structure, in which a layer of dynamic, partially unfolded and extended proteins surrounds a micelle-like fatty acid core. This structure, first reported for α-lactalbumin together with OA, resembles complexes formed between proteins and anionic surfactants like SDS. Liprotides first rose to fame through their anti-carcinogenic properties which still remains promising for topical applications though not yet implemented in the clinic. In addition, liprotides show potential in drug delivery thanks to the ability of the micelle core to solubilize and stabilize hydrophobic compounds, though applications are challenged by their sensitivity to acidic pH and dynamic exchange of lipids which makes them easy prey for serum "hoovers" such as albumin. However, liprotides are also of fundamental interest as a generic "protein complex structure", demonstrating the many and varied structural consequences of protein-lipid interactions. Here we provide an overview of the different types of liprotide complexes, ranging from quasi-native complexes via core-shell structures to multi-layer structures, and discuss the many conditions under which they form. Given the many variable types of complexes that can form, rigorous biophysical analysis (stoichiometry, shape and structure of the complexes) remains crucial for a complete understanding of the mechanisms of action of this fascinating group of protein-lipid complexes both in vitro and in vivo.
Collapse
Affiliation(s)
- Jannik Nedergaard Pedersen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Henriette Kristina Søster Frislev
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; Novo Nordisk, Hallas Alle 1, DK-4400 Kalundborg, Denmark
| | - Jan Skov Pedersen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; Department of Chemistry, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Daniel Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark.
| |
Collapse
|
11
|
El-Fakharany EM, Redwan EM. Protein-lipid complexes: molecular structure, current scenarios and mechanisms of cytotoxicity. RSC Adv 2019; 9:36890-36906. [PMID: 35539089 PMCID: PMC9075609 DOI: 10.1039/c9ra07127j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/21/2019] [Indexed: 02/04/2023] Open
Abstract
Some natural proteins can be complexed with oleic acid (OA) to form an active protein-lipid formulation that can induce tumor-selective apoptosis. The first explored protein was human milk α-lactalbumin (α-LA), called HAMLET when composed with OA in antitumor form. Several groups have prepared active protein-lipid complexes using a variety of approaches, all of which depend on target protein destabilization or direct OA-protein incubation to alter pH to acid or alkaline condition. In addition to performing vital roles in inflammatory processes and immune responses, fatty acids can disturb different metabolic pathways and cellular signals. Therefore, the tumoricidal action of these complexes is related to OA rather than the protein that keeps OA in solution and acts as a vehicle for transferring OA molecules to tumor cells. However, other studies have suggested that the antitumor efficacy of these complexes was exerted by both protein and OA together. The potential is not limited to the anti-tumor activity of protein-lipid complexes but extends to other functions such as bactericidal activity. The protein shell enhances the solubility and stability of the bound fatty acid. These protein-lipid complexes are promising candidates for fighting various cancer types and managing bacterial and viral infections.
Collapse
Affiliation(s)
- Esmail M El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications (SRTA-City) New Borg EL-Arab 21934 Alexandria Egypt
| | - Elrashdy M Redwan
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications (SRTA-City) New Borg EL-Arab 21934 Alexandria Egypt
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University P. O. Box 80203 Jeddah Saudi Arabia
| |
Collapse
|
12
|
Nutritional and therapeutic perspectives of camel milk and its protein hydrolysates: A review on versatile biofunctional properties. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103441] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
13
|
Abstract
Oleic acid (OA) is a monounsaturated fatty acid that upon binding to milk proteins, such as α-lactalbumin and lactoferrin, forms potent complexes, which exert selective anti-tumor activity against malignant cells but are nontoxic for healthy normal cells. We showed that the interaction of OA with albumins isolated from human, bovine, and camel milk results in the formation of complexes with high antitumor activity against Caco-2, HepG-2, PC-3, and MCF-7 tumor cells. The antitumor effect of the complexes is mostly due to the action of oleic acid, similar to the case of OA complexes with other proteins. Viability of tumor cells is inhibited by the albumin-OA complexes in a dose dependent manner, as evaluated by the MTT assay. Strong induction of apoptosis in tumor cells after their treatment with the complexes was monitored by flow cytometry, cell cycle analysis, nuclear staining, and DNA fragmentation methods. The complex of camel albumin with OA displayed the most pronounced anti-tumor effects in comparison with the complexes of OA with human and bovine albumins. Therefore, these results suggest that albumins have the potential to be used as efficient and low cost means of tumor treatment.
Collapse
|
14
|
Krajewska M, Dopierała K, Weiss M, Prochaska K. Temperature, pH, and Molecular Packing Effects on the Penetration of Oleic Acid Monolayer by α-Lactalbumin. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:3183-3193. [PMID: 30706714 DOI: 10.1021/acs.langmuir.8b04153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Recently, we reported on the interfacial behavior of mixed oleic acid (OA)-α-lactalbumin monolayer and its relevance in the formation of tumoricidal HAMLET (human α-lactalbumin made lethal to tumor cells)-like complex. This complex is probably formed in the gastrointestinal tract, but it has not been proved so far. The molecular base and the underlying physicochemical forces leading to such complexation remain to be known as well. There are also several other issues related with the complex stoichiometry that need to be fully explained. This study provides insight into the mechanism of temperature, pH, and physical state of monolayer-dependent binding of OA by the milk protein- apo-α-lactalbumin. Using the Langmuir and Langmuir-Blodgett approaches, we investigated the interactions between the OA monolayer and the apo-bovine α-lactalbumin (BLA III) at different pH, temperatures, and molecular packing. We found that the most favorable conditions for the formation of mixed OA-BLA III film are relevant to the gastric environment. The stabilization of OA-BLA III at the interface is associated with the conformational changes of protein in the presence of fatty acids induced by low pH and high temperature in the expanded monolayer. Our approach helps to understand the molecular mechanism of HAMLET/bovine α-lactalbumin made lethal to tumor cells formation in vivo.
Collapse
Affiliation(s)
- Martyna Krajewska
- Institute of Chemical Technology and Engineering , Poznan University of Technology , Berdychowo 4 , 60-965 Poznań , Poland
| | - Katarzyna Dopierała
- Institute of Chemical Technology and Engineering , Poznan University of Technology , Berdychowo 4 , 60-965 Poznań , Poland
| | - Marek Weiss
- Institute of Physics , Poznan University of Technology , Piotrowo 3 , 60-965 Poznań , Poland
| | - Krystyna Prochaska
- Institute of Chemical Technology and Engineering , Poznan University of Technology , Berdychowo 4 , 60-965 Poznań , Poland
| |
Collapse
|
15
|
Park Y, Park Y, Jin S, Kim JW, Jung YM. Formation mechanism of BAMLET by 2D Raman correlation analysis. J Mol Struct 2018. [DOI: 10.1016/j.molstruc.2018.05.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
16
|
Abstract
α-Lactalbumin is a whey protein that constitutes approximately 22% of the proteins in human milk and approximately 3.5% of those in bovine milk. Within the mammary gland, α-lactalbumin plays a central role in milk production as part of the lactose synthase complex required for lactose formation, which drives milk volume. It is an important source of bioactive peptides and essential amino acids, including tryptophan, lysine, branched-chain amino acids, and sulfur-containing amino acids, all of which are crucial for infant nutrition. α-Lactalbumin contributes to infant development, and the commercial availability of α-lactalbumin allows infant formulas to be reformulated to have a reduced protein content. Likewise, because of its physical characteristics, which include water solubility and heat stability, α-lactalbumin has the potential to be added to food products as a supplemental protein. It also has potential as a nutritional supplement to support neurological function and sleep in adults, owing to its unique tryptophan content. Other components of α-lactalbumin that may have usefulness in nutritional supplements include the branched-chain amino acid leucine, which promotes protein accretion in skeletal muscle, and bioactive peptides, which possess prebiotic and antibacterial properties. This review describes the characteristics of α-lactalbumin and examines the potential applications of α-lactalbumin for human health.
Collapse
Affiliation(s)
- Donald K Layman
- Department of Food Science and Human Nutrition, University of Illinois at Urban-Champaign, Urbana, Illinois, USA
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, California, USA
| | - John D Fernstrom
- Department of Psychiatry and the Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
17
|
Breydo L, Almehdar HA, El-Fakharany EM, Redwan EM, Uversky VN. Not all AMLETs are made equal: complexes of cow and camel α-lactalbumin with oleic acid show different structure and stability. J Biomol Struct Dyn 2017; 36:4405-4412. [PMID: 29237329 DOI: 10.1080/07391102.2017.1417163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Leonid Breydo
- a Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute , Morsani College of Medicine, University of South Florida , Tampa 33612 , FL , USA
| | - Hussein A Almehdar
- b Department of Biological Science, Faculty of Science , King Abdulaziz University , PO Box 80203, Jeddah 21589 , Saudi Arabia
| | - Esmail M El-Fakharany
- c Therapeutic and Protective Proteins Laboratory, Protein Research Department , Genetic Engineering and Biotechnology Research Institute GEBRI, City for Scientific Research and Technology Applications , New Borg EL Arab, Alexandria 21934 , Egypt
| | - Elrashdy M Redwan
- b Department of Biological Science, Faculty of Science , King Abdulaziz University , PO Box 80203, Jeddah 21589 , Saudi Arabia.,c Therapeutic and Protective Proteins Laboratory, Protein Research Department , Genetic Engineering and Biotechnology Research Institute GEBRI, City for Scientific Research and Technology Applications , New Borg EL Arab, Alexandria 21934 , Egypt
| | - Vladimir N Uversky
- a Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute , Morsani College of Medicine, University of South Florida , Tampa 33612 , FL , USA.,b Department of Biological Science, Faculty of Science , King Abdulaziz University , PO Box 80203, Jeddah 21589 , Saudi Arabia.,d Laboratory of New Methods in Biology , Institute for Biological Instrumentation of the Russian Academy of Sciences , Pushchino 142290 , Moscow region , Russia
| |
Collapse
|
18
|
Uversky VN, El-Fakharany EM, Abu-Serie MM, Almehdar HA, Redwan EM. Divergent Anticancer Activity of Free and Formulated Camel Milk α-Lactalbumin. Cancer Invest 2017; 35:610-623. [PMID: 28949782 DOI: 10.1080/07357907.2017.1373783] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Alpha-lactalbumin (α-LA), a small milk calcium-binding globular protein, is known to possess noticeable anticancer activity, which is determined by the ability of this protein to form complexes with oleic acid (OA). To date, in addition to human and bovine α-LA, the ability to form such anti-tumor complexes with OA was described for goat and camel α-LA. Although the mechanisms of the anticancer activity of human and bovine α-LA are already well-studied, little is currently known about the anticancer action of this camel protein. The goal of this study was to fill this gap and to analyze the anticancer and pro-apoptotic activities of camel α-LA in its free form (α-cLA) and as an OA-containing complex (OA-α-cLA) using four human cancer cell lines, including Caco-2 colon cancer cells, PC-3 prostate cancer cells, HepG-2 hepatoma cells, and MCF-7 breast cancer cells as targets. The anti-tumor activities of OA-α-cLA and α-cLA were analyzed using MTT test, annexin/PI staining, cell cycle analysis, nuclear staining, and tyrosine kinase (TK) inhibition methods. We show here that the OA-α-cLA complex does not affect normal cells but has noticeable anti-cancer activity, especially against MCF-7 cells, thus boosting the anticancer activity of α-cLA and improving the selectivity of OA. The OA-α-cLA complex mediated cancer cell death via selective induction of apoptosis and cell-cycle arrest at lower IC50 than that of free α-cLA by more than two folds. However, OA induced apoptosis at higher extent than OA-α-cLA and α-cLA. OA also caused unselective apoptosis-dependent cell death in both normal and cancer cells to a similar degree. The apoptosis and cell-cycle arresting effect of OA-α-cLA may be attributed to the TK inhibition activity of OA. Therefore, OA-α-cLA serves as efficient anticancer complex with two functional components, α-cLA and OA, possessing different activities. This study declared the effectiveness of OA-α-cLA complex as a promising entity with anticancer activity, and these formulated OA-camel protein complexes constitute an auspicious approach for cancer remedy, particularly for breast cancer.
Collapse
Affiliation(s)
- Vladimir N Uversky
- a Department of Biological Sciences, Faculty of Sciences , King Abdulaziz University , Jeddah , Saudi Arabia.,b Institute for Biological Instrumentation of the Russian Academy of Sciences , Pushchino , Moscow region , Russia.,c Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute , Morsani College of Medicine, University of South Florida , Tampa , FL , USA
| | - Esmail M El-Fakharany
- d Therapeutic and Protective Proteins Laboratory, Protein Research Department, Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute , City for Scientific Research and Technology Applications (SRTA-City) , Alexandria , Egypt
| | - Marwa M Abu-Serie
- e Medical Biotechnology Department , Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications (SRTA-City) , Alexandria , Egypt
| | - Hussein A Almehdar
- a Department of Biological Sciences, Faculty of Sciences , King Abdulaziz University , Jeddah , Saudi Arabia
| | - Elrashdy M Redwan
- a Department of Biological Sciences, Faculty of Sciences , King Abdulaziz University , Jeddah , Saudi Arabia.,d Therapeutic and Protective Proteins Laboratory, Protein Research Department, Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute , City for Scientific Research and Technology Applications (SRTA-City) , Alexandria , Egypt
| |
Collapse
|
19
|
Chaudhuri A, Prasanna X, Agiru P, Chakraborty H, Rydström A, Ho JCS, Svanborg C, Sengupta D, Chattopadhyay A. Protein-dependent Membrane Interaction of A Partially Disordered Protein Complex with Oleic Acid: Implications for Cancer Lipidomics. Sci Rep 2016; 6:35015. [PMID: 27731329 PMCID: PMC5059734 DOI: 10.1038/srep35015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/09/2016] [Indexed: 12/18/2022] Open
Abstract
Bovine α-lactalbumin (BLA) forms cytotoxic complexes with oleic acid (OA) that perturbs tumor cell membranes, but molecular determinants of these membrane-interactions remain poorly understood. Here, we aim to obtain molecular insights into the interaction of BLA/BLA-OA complex with model membranes. We characterized the folding state of BLA-OA complex using tryptophan fluorescence and resolved residue-specific interactions of BLA with OA using molecular dynamics simulation. We integrated membrane-binding data using a voltage-sensitive probe and molecular dynamics (MD) to demonstrate the preferential interaction of the BLA-OA complex with negatively charged membranes. We identified amino acid residues of BLA and BLA-OA complex as determinants of these membrane interactions using MD, functionally corroborated by uptake of the corresponding α-LA peptides across tumor cell membranes. The results suggest that the α-LA component of these cytotoxic complexes confers specificity for tumor cell membranes through protein interactions that are maintained even in the lipid complex, in the presence of OA.
Collapse
Affiliation(s)
- Arunima Chaudhuri
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | | | - Priyanka Agiru
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Hirak Chakraborty
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Anna Rydström
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - James C. S. Ho
- Centre for Biomimetic Sensor Science, School of Materials Science & Engineering, Nanyang Technological University, 637553 Singapore
| | - Catharina Svanborg
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | | | | |
Collapse
|
20
|
Jung S, Lee S, Lee H, Yoon J, Lee EK. Oleic acid-embedded nanoliposome as a selective tumoricidal agent. Colloids Surf B Biointerfaces 2016; 146:585-9. [PMID: 27424089 DOI: 10.1016/j.colsurfb.2016.06.058] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/20/2022]
Abstract
HAMLET (Human Alpha-lactalbumin Made LEthal to Tumor cell), a molecular complex of human α-lactalbumin and oleic acid, is known to have selective cytotoxic activity against certain types of tumors. This cytotoxicity is known to stem from water-insoluble oleic acid. In this study, we manufactured an alternative complex using liposome as an oleic acid delivery vesicle. We named this nanolipoplex LIMLET (LIposome Made LEthal to Tumor cell). The LIMLET vesicle contained approximately 90,200 oleic acid molecules inserted into its lipophilic phospholipid bilayer and had a nominal mean diameter of 127nm. Using a WST-1 assay, its cytotoxicity against two cancer cell lines, MDA-MB-231 (human breast cancer) and A549 (human lung cancer), were tested. The results were compared with that of a normal cell line, Vero (from monkey kidney). We found that (1) LIMLET showed distinctive cytotoxicity against A549 and MDA-MB-231 cells, whereas bare liposomes (containing no oleic acid) had no toxicity, even at high concentrations, and (2) LIMLET demonstrated selective, concentration-dependent toxicity against the cancer cells: the LD50 values of MDA-MB-231 and A549 cells were 1.3 and 2.2nM LIMLET, respectively, whereas the LD50 of Vero was 5.7nM. The strength of the tumoricidal effect appeared to stem from the number of oleic acid molecules present. Our result suggests that LIMLET, like HAMLET, is an interesting nanolipoplex that can potentially be developed into tumor treatments.
Collapse
Affiliation(s)
- Sujin Jung
- Department of Bionano Engineering, Hanyang University-ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Sangah Lee
- Department of Bionano Engineering, Hanyang University-ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Hyejin Lee
- Department of Bionano Engineering, Hanyang University-ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Jaejin Yoon
- Department of Bionano Engineering, Hanyang University-ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - E K Lee
- Department of Bionano Engineering, Hanyang University-ERICA, Ansan, Gyeonggi-do 15588, Republic of Korea.
| |
Collapse
|
21
|
Hoque M, Gupta J, Rabbani G, Khan RH, Saleemuddin M. Behaviour of oleic acid-depleted bovine alpha-lactalbumin made LEthal to tumor cells (BAMLET). MOLECULAR BIOSYSTEMS 2016; 12:1871-80. [DOI: 10.1039/c5mb00905g] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Oleic acid (OA) complexes of human alpha-lactalbumin (α-LA) and several other proteins are effective in the killing of a variety of tumor cells.
Collapse
Affiliation(s)
- Mehboob Hoque
- Interdisciplinary Biotechnology Unit
- Aligarh Muslim University
- Aligarh 202002
- India
| | - Jyoti Gupta
- Interdisciplinary Biotechnology Unit
- Aligarh Muslim University
- Aligarh 202002
- India
| | - Gulam Rabbani
- Interdisciplinary Biotechnology Unit
- Aligarh Muslim University
- Aligarh 202002
- India
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit
- Aligarh Muslim University
- Aligarh 202002
- India
| | - M. Saleemuddin
- Interdisciplinary Biotechnology Unit
- Aligarh Muslim University
- Aligarh 202002
- India
| |
Collapse
|
22
|
Mahanta S, Paul S. Stable Self-Assembly of Bovine α-Lactalbumin Exhibits Target-Specific Antiproliferative Activity in Multiple Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:28177-28187. [PMID: 26440360 DOI: 10.1021/acsami.5b06076] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Self-assembly of a protein is a natural phenomenon; however, the process can be performed under a suitable condition in vitro. Since proteins are nontoxic, biodegradable, and biocompatible in nature, they are used in various industrial applications such as biocatalyst, therapeutic agent, and drug carriers. Moreover, their flexible structural state and specific activity are being used as sensors and immensely attract many new applications. However, the inherent potential of protein self-assembly for various applications is yet to be explored in detail. In this study, spherical self-assembly of bovine α-lactalbumin (nsBLA) was synthesized using an optimized ethanol-mediated desolvation process with an average diameter of approximately 300 nm. The self-assembly was found to be highly stable against thermal, pH, and proteases stress. When nsBLA was administered in various cancer cells, it demonstrated high cytotoxicity in three different cancer cells via reactive oxygen species (ROS) generation, whereas it exhibited negligible toxicity in normal human and murine cells. When nsBLA was conjugated with folic acid, it improved the cytotoxicity and perhaps mediated through enhanced cellular uptake in cancer cells through binding with folate receptors. Further, experimental results confirmed that the cancer cell death induced by nsBLA was not caused by apoptosis but a necrotic-like death mechanism. When compared with a well-known protein-based anticancer agent BAMLET (bovine α-lactalbumin made lethal against tumor cell), the self-assembled BLA clearly exhibited higher cytotoxicity to cancer cells than BAMLET. While BAMLET exhibits poor biocompatibility, our nsBLA demonstrated excellent biocompatibility to normal cells. Therefore, in this study, we prepared self-assembled α-lactalbumin that exhibits strong inherent antiproliferative potential in multiple cancer cells which can be used for efficient therapeutic approach in cancer.
Collapse
Affiliation(s)
- Sailendra Mahanta
- Structural Biology and Nanomedicine Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology , Rourkela 769008, Odisha, India
| | - Subhankar Paul
- Structural Biology and Nanomedicine Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology , Rourkela 769008, Odisha, India
| |
Collapse
|
23
|
Nongonierma AB, FitzGerald RJ. Bioactive properties of milk proteins in humans: A review. Peptides 2015; 73:20-34. [PMID: 26297879 DOI: 10.1016/j.peptides.2015.08.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/13/2015] [Accepted: 08/14/2015] [Indexed: 12/25/2022]
Abstract
Many studies have demonstrated that milk protein consumption has benefits in terms of promoting human health. This review assesses the intervention studies which have evaluated potential health enhancing effects in humans following the ingestion of milk proteins. The impact of milk protein ingestion has been studied to asses their satiating, hypotensive, antimicrobial, anti-inflammatory, anticancer, antioxidant and insulinotropic properties as well as their impact on morphological modifications (e.g., muscle and fat mass) in humans. Consistent health promoting effects appear to have been observed in certain instances (i.e., muscle protein synthesis, insulinotropic and hypotensive activity). However, controversial outcomes have also been reported (i.e., antimicrobial, anti-inflammatory, anticancer and antioxidant properties). Several factors including interindividual differences, the timing of protein ingestion as well as the potency of the active components may explain these differences. In addition, processing conditions have been reported, in certain instances, to affect milk protein structure and therefore modify their bioactive potential. It is thought that the health promoting properties of milk proteins are linked to the release of bioactive peptides (BAPs) during gastrointestinal digestion. There is a need for further research to develop a more in-depth understanding on the possible mechanisms involved in the observed physiological effects. In addition, more carefully controlled and appropriately powered human intervention studies are required to demonstrate the health enhancing properties of milk proteins in humans.
Collapse
Affiliation(s)
- Alice B Nongonierma
- Department of Life Sciences and Food for Health Ireland (FHI), University of Limerick, Limerick, Ireland
| | - Richard J FitzGerald
- Department of Life Sciences and Food for Health Ireland (FHI), University of Limerick, Limerick, Ireland.
| |
Collapse
|
24
|
Wen H, Strømland Ø, Halskau Ø. α-Lactalbumin:Oleic Acid Complex Spontaneously Delivers Oleic Acid to Artificial and Erythrocyte Membranes. J Mol Biol 2015; 427:3177-87. [PMID: 26297199 DOI: 10.1016/j.jmb.2015.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/08/2015] [Indexed: 10/23/2022]
Abstract
Human α-lactalbumin made lethal to tumor cells (HAMLET) is a tumoricidal complex consisting of human α-lactalbumin and multiple oleic acids (OAs). OA has been shown to play a key role in the activity of HAMLET and its related complexes, generally known as protein-fatty acid (PFA) complexes. In contrast to what is known about the fate of the protein component of such complexes, information about what happens to OA during their action is still lacking. We monitored the membrane, OA and protein components of bovine α-lactalbumin complexed with OA (BLAOA; a HAMLET-like substance) and how they associate with each other. Using ultracentrifugation, we found that the OA and lipid components follow each other closely. We then firmly identify a transfer of OA from BLAOA to both artificial and erythrocyte membranes, indicating that natural cells respond similarly to BLAOA treatment as artificial membranes. Uncomplexed OA is unable to similarly affect membranes at the conditions tested, even at elevated concentrations. Thus, BLAOA can spontaneously transfer OA to a lipid membrane. After the interaction with the membrane, the protein is likely to have lost most or all of its OA. We suggest a mechanism for passive import of mainly uncomplexed protein into cells, using existing models for OA's effect on membranes. Our results are consistent with a membrane destabilization mediated predominantly by OA insertion being a significant contribution to PFA cytotoxicity.
Collapse
Affiliation(s)
- Hanzhen Wen
- Department of Molecular Biology, University of Bergen, Thormøhlens Gate 55, 5008 Bergen, Norway
| | - Øyvind Strømland
- Department of Molecular Biology, University of Bergen, Thormøhlens Gate 55, 5008 Bergen, Norway
| | - Øyvind Halskau
- Department of Molecular Biology, University of Bergen, Thormøhlens Gate 55, 5008 Bergen, Norway.
| |
Collapse
|
25
|
Nongonierma AB, FitzGerald RJ. The scientific evidence for the role of milk protein-derived bioactive peptides in humans: A Review. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.06.021] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
26
|
Delgado Y, Morales-Cruz M, Figueroa CM, Hernández-Román J, Hernández G, Griebenow K. The cytotoxicity of BAMLET complexes is due to oleic acid and independent of the α-lactalbumin component. FEBS Open Bio 2015; 5:397-404. [PMID: 26101738 PMCID: PMC4430638 DOI: 10.1016/j.fob.2015.04.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/16/2015] [Indexed: 11/17/2022] Open
Abstract
We synthesized three different BAMLET complexes consisting of oleic acid coupled to bovine α-lactalbumin. Oleic acid micelles alone are tumoricidal at equimolar concentrations of oleic acid bound in the BAMLET complexes. α-Lactalbumin is non-toxic to cells even when delivered to their cytoplasm. Both, BAMLET and oleic acid micelles showed no selective cytotoxicity to cancer cells.
Lipid–protein complexes comprised of oleic acid (OA) non-covalently coupled to human/bovine α-lactalbumin, named HAMLET/BAMLET, display cytotoxic properties against cancer cells. However, there is still a substantial debate about the role of the protein in these complexes. To shed light into this, we obtained three different BAMLET complexes using varying synthesis conditions. Our data suggest that to form active BAMLET particles, OA has to reach critical micelle concentration with an approximate diameter of 250 nm. Proteolysis experiments on BAMLET show that OA protects the protein and is probably located on the surface, consistent with a micelle-like structure. Native or unfolded α-lactalbumin without OA lacked any tumoricidal activity. In contrast, OA alone killed cancer cells with the same efficiency at equimolar concentrations as its formulation as BAMLET. Our data show unequivocally that the cytotoxicity of the BAMLET complex is exclusively due to OA and that OA alone, when formulated as a micelle, is as toxic as the BAMLET complex. The contradictory literature results on the cytotoxicity of BAMLET might be explained by our finding that it was imperative to sonicate the samples to obtain toxic OA.
Collapse
Key Words
- BAMLET
- BAMLET, bovine α-lactalbumin made lethal to tumor cells
- Cancer therapy
- DLS, dynamic light scattering
- EPR, enhanced permeability and retention
- FA, fatty acid
- Fatty acid
- FoA, folic acid
- HAMLET
- HAMLET, human α-lactalbumin made lethal to tumor cells
- MTS, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium
- NP, nanoparticles
- OA, oleic acid
- Oleic acid
- PMS, phenazine methosulfate
- SEM, scanning electron microscopy
- α-LA, α-lactalbumin
- α-Lactalbumin
Collapse
Affiliation(s)
- Yamixa Delgado
- Department of Biology, University of Puerto Rico, Río Piedras Campus, P.O. Box 23360, San Juan 00931-3346, Puerto Rico
| | - Moraima Morales-Cruz
- Department of Biology, University of Puerto Rico, Río Piedras Campus, P.O. Box 23360, San Juan 00931-3346, Puerto Rico
| | - Cindy M. Figueroa
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, P.O. Box 23360, San Juan 00931-3346, Puerto Rico
| | - José Hernández-Román
- Department of Biology, University of Puerto Rico, Río Piedras Campus, P.O. Box 23360, San Juan 00931-3346, Puerto Rico
| | - Glinda Hernández
- Department of Biology, University of Puerto Rico, Río Piedras Campus, P.O. Box 23360, San Juan 00931-3346, Puerto Rico
| | - Kai Griebenow
- Department of Biology, University of Puerto Rico, Río Piedras Campus, P.O. Box 23360, San Juan 00931-3346, Puerto Rico
- Department of Chemistry, University of Puerto Rico, Río Piedras Campus, P.O. Box 23360, San Juan 00931-3346, Puerto Rico
- Corresponding author at: Department of Chemistry, University of Puerto Rico, Río Piedras Campus, P.O. Box 23360, San Juan 00931-3346, Puerto Rico. Tel.: +1 (787) 764 0000x7374; fax: +1 (787) 756 8242.
| |
Collapse
|
27
|
Hoque M, Nanduri R, Gupta J, Mahajan S, Gupta P, Saleemuddin M. Oleic acid complex of bovine α-lactalbumin induces eryptosis in human and other erythrocytes by a Ca(2+)-independent mechanism. Biochim Biophys Acta Gen Subj 2015; 1850:1729-39. [PMID: 25913522 DOI: 10.1016/j.bbagen.2015.04.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/08/2015] [Accepted: 04/16/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Complexes of oleic acid (OA) with milk α-lactalbumin, received remarkable attention in view of their selective toxicity towards a spectrum of tumors during the last two decades. OA complexes of some structurally related/unrelated proteins are also tumoricidal. Erythrocytes are among the few differentiated cells that are sensitive and undergo hemolysis when exposed to the complexes. METHODS The effects of OA complex of bovine α-lactalbumin (Bovine Alpha-lactalbumin Made LEthal to Tumor cells, BAMLET) on human, goat and chicken erythrocytes on calcein leakage, phosphatidylserine exposure, morphological changes and hemolysis were studied by confocal microscopy, FACS analysis, scanning electron microscopy and measuring hemoglobin release. RESULTS Erythrocytes exposed to BAMLET undergo eryptosis-like alterations as revealed by calcein leakage, surface phosphatidylserine exposure and transformation to echinocytes at low concentrations and hemolysis when the concentration of the complex was raised. Ca(2+) was not essential and restricted the alterations when included in the medium. The BAMLET-induced alterations in human erythrocytes were prevented by the cation channel inhibitors, amiloride and BaCl2 but not by inhibitors of thiol proteases, sphingomyelinase and by the antioxidant N-acetyl cysteine. CONCLUSIONS The work shows for the first time that low concentrations of BAMLET induces eryptosis in erythrocytes by a novel mechanism not requiring Ca(2+) and hemolysis by detergent-like action by the released OA at higher concentrations. GENERAL SIGNIFICANCE The study points out to the need for a comprehensive evaluation of the toxicity of OA complexes of α-lactalbumin and other proteins towards erythrocytes and other differentiated cells before being considered for therapy.
Collapse
Affiliation(s)
- Mehboob Hoque
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | | | - Jyoti Gupta
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Sahil Mahajan
- CSIR-Institute of Microbial Technology, Chandigarh 160036, India
| | - Pawan Gupta
- CSIR-Institute of Microbial Technology, Chandigarh 160036, India
| | - M Saleemuddin
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
28
|
Lezhnin YN, Kravchenko YE, Frolova EI, Chumakov PM, Chumakov SP. Oncotoxic proteins in cancer therapy: Mechanisms of action. Mol Biol 2015. [DOI: 10.1134/s0026893315020077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Fang B, Zhang M, Tian M, Ren FZ. Self-assembled β-lactoglobulin-oleic acid and β-lactoglobulin-linoleic acid complexes with antitumor activities. J Dairy Sci 2015; 98:2898-907. [PMID: 25771044 DOI: 10.3168/jds.2014-8993] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 01/23/2015] [Indexed: 11/19/2022]
Abstract
β-Lactoglobulin (β-LG) can bind to fatty acids such as oleic acid (OA) and linoleic acid (LA). Another whey protein, α-lactalbumin (α-LA), can also bind to OA to give the complex α-LA-OA, which has antitumor properties. Based on reports that the activity of α-LA-OA is highly dependent on OA, as well as the acquisition of similar complexes using other proteins, such as lysozyme and lactoferrin, we speculated whether β-LG could also kill tumor cells after binding to other fatty acids. Therefore, we prepared complexes of β-LG with OA (β-LG-OA) and LA (β-LG-LA) in the current study and evaluated them in terms of antitumor activity and thermostability using the methylene blue method and differential scanning calorimetry, respectively. The structural features of these complexes were also evaluated using fluorescence spectroscopy and circular dichroism. The binding dynamics of OA and LA to β-LG were studied using isothermal titration calorimetry. Cell viability results revealed that β-LG-LA and β-LG-OA exhibited similar antitumor activities. Interestingly, the binding of β-LG to LA led to an increase in its thermostability, whereas its binding to OA had very little effect. The environments of the tryptophan residues in the β-LG-OA and β-LG-LA complexes were very different, with the residues being blue- and red-shifted, respectively. Furthermore, the hydrophobic regions in β-LG were buried after binding of OA, which was slightly changed in β-LG-LA. Circular dichroism results showed that β-LG-OA enhanced the tertiary structure, which was partially lost in β-LG-LA. There were more binding sites for OA than for LA on β-LG, although the binding constants of the 2 fatty acids were similar, with both acids interacting with the protein though van der Waals and hydrogen bonding interactions. This study could help provide a deeper understanding of the structural basis for formation of antitumor protein-fatty acid complexes.
Collapse
Affiliation(s)
- B Fang
- Beijing Laboratory for Food Quality and Safety, China Agricultural University, Beijing 100083, China; Academy of State Administration of Grain, Beijing 100037, China
| | - M Zhang
- Beijing Technology and Business University, School of Food Science and Chemical Engineering, Beijing 100048, China
| | - M Tian
- Academy of State Administration of Grain, Beijing 100037, China
| | - F Z Ren
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
30
|
Chung SY, Mattison CP, Reed S, Wasserman RL, Desormeaux WA. Treatment with oleic acid reduces IgE binding to peanut and cashew allergens. Food Chem 2015; 180:295-300. [PMID: 25766831 DOI: 10.1016/j.foodchem.2015.02.056] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/26/2015] [Accepted: 02/12/2015] [Indexed: 01/03/2023]
Abstract
Oleic acid (OA) is known to bind and change the bioactivities of proteins, such as α-lactalbumin and β-lactoglobulin in vitro. The objective of this study was to determine if OA binds to allergens from a peanut extract or cashew allergen and changes their allergenic properties. Peanut extract or cashew allergen (Ana o 2) was treated with or without 5mM sodium oleate at 70°C for 60 min (T1) or under the same conditions with an additional overnight incubation at 37°C (T2). After treatment, the samples were dialyzed and analyzed by SDS-PAGE and for OA content. IgE binding was evaluated by ELISA and western blot, using a pooled serum or plasma from individuals with peanut or cashew allergies. Results showed that OA at a concentration of 5mM reduced IgE binding to the allergens. Peanut sample T2 exhibited a lower IgE binding and a higher OA content (protein-bound) than T1. Cashew allergen T2 also showed a reduction in IgE binding. We conclude that OA reduces the allergenic properties of peanut extract and cashew allergen by binding to the allergens. Our findings indicate that OA in the form of sodium oleate may be potentially useful as a coating to reduce the allergenic properties of peanut and cashew allergens.
Collapse
Affiliation(s)
- Si-Yin Chung
- USDA-ARS, Southern Regional Research Center, New Orleans, LA, USA.
| | | | - Shawndrika Reed
- USDA-ARS, Southern Regional Research Center, New Orleans, LA, USA
| | - Richard L Wasserman
- Allergy Immunology Research Center of North Texas, Department of Pediatrics, University of Texas Southwestern Medical School, Dallas, TX 75390, USA
| | | |
Collapse
|
31
|
Kaspersen JD, Pedersen JN, Hansted JG, Nielsen SB, Sakthivel S, Wilhelm K, Nemashkalova EL, Permyakov SE, Permyakov EA, Pinto Oliveira CL, Morozova-Roche LA, Otzen DE, Pedersen JS. Generic structures of cytotoxic liprotides: nano-sized complexes with oleic acid cores and shells of disordered proteins. Chembiochem 2014; 15:2693-702. [PMID: 25403886 DOI: 10.1002/cbic.201402407] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Indexed: 11/05/2022]
Abstract
The cytotoxic complex formed between α-lactalbumin and oleic acid (OA) has inspired many studies on protein-fatty acid complexes, but structural insight remains sparse. After having used small-angle X-ray scattering (SAXS) to obtain structural information, we present a new, generic structural model of cytotoxic protein-oleic acid complexes, which we have termed liprotides (lipids and partially denatured proteins). Twelve liprotides formed from seven structurally unrelated proteins and prepared by different procedures all displayed core-shell structures, each with a micellar OA core and a shell consisting of flexible, partially unfolded protein, which stabilizes the OA micelle. The common structure explains similar effects exerted on cells by different liprotides and is consistent with a cargo off-loading of the OA into cell membranes.
Collapse
Affiliation(s)
- Jørn D Kaspersen
- Department of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C (Denmark)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Lactaptin induces p53-independent cell death associated with features of apoptosis and autophagy and delays growth of breast cancer cells in mouse xenografts. PLoS One 2014; 9:e93921. [PMID: 24710119 PMCID: PMC3978064 DOI: 10.1371/journal.pone.0093921] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 03/11/2014] [Indexed: 11/19/2022] Open
Abstract
Lactaptin, the proteolytic fragment of human milk kappa-casein, induces the death of various cultured cancer cells. The mechanisms leading to cell death after lactaptin treatment have not been well characterized. In this study the in vivo and in vitro effects of a recombinant analogue of lactaptin (RL2) were examined. Following treatment with the recombinant analogue of lactaptin strong caspase -3, -7 activation was detected. As a consequence of caspase activation we observed the appearance of a sub-G1 population of cells with subdiploid DNA content. Dynamic changes in the mRNA and protein levels of apoptosis-related genes were estimated. No statistically reliable differences in p53 mRNA level or protein level were found between control and RL2-treated cells. We observed that RL2 constitutively suppressed bcl-2 mRNA expression and down regulated Bcl-2 protein expression in MDA-MB-231 cells. We demonstrated that RL2 penetrates cancer and non-transformed cells. Identification of the cellular targets of the lactaptin analogue revealed that α/β-tubulin and α-actinin-1 were RL2-bound proteins. As the alteration in cellular viability in response to protein stimulus can be realized not only by way of apoptosis but also by autophagy, we examined the implications of autophagy in RL2-dependent cell death. We also found that RL2 treatment induces LC3-processing, which is a hallmark of autophagy. The autophagy inhibitor chloroquine enhanced RL2 cytotoxicity to MDA-MB-231 cells, indicating the pro-survival effect of RL2-dependent autophagy. The antitumour potential of RL2 was investigated in vivo in mouse xenografts bearing MDA-MB-231 cells. We demonstrated that the recombinant analogue of lactaptin significantly suppressed the growth of solid tumours. Our results indicate that lactaptin could be a new molecule for the development of anticancer drugs.
Collapse
|
33
|
Rath EM, Duff AP, Håkansson AP, Knott RB, Church WB. Small-angle X-ray scattering of BAMLET at pH 12: a complex of α-lactalbumin and oleic acid. Proteins 2014; 82:1400-8. [PMID: 24408789 DOI: 10.1002/prot.24508] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 12/04/2013] [Accepted: 12/26/2013] [Indexed: 11/11/2022]
Abstract
BAMLET (Bovine Alpha-lactalbumin Made LEthal to Tumors) is a member of the family of the HAMLET-like complexes, a novel class of protein-based anti-cancer complexes that incorporate oleic acid and deliver it to cancer cells. Small angle X-ray scattering (SAXS) was performed on the complex at pH 12, examining the high pH structure as a function of oleic acid added. The SAXS data for BAMLET species prepared with a range of oleic acid concentrations indicate extended, irregular, partially unfolded protein conformations that vary with the oleic acid concentration. Increases in oleic acid concentration correlate with increasing radius of gyration without an increase in maximum particle dimension, indicating decreasing protein density. The models for the highest oleic acid content BAMLET indicate an unusual coiled elongated structure that contrasts with apo-α-lactalbumin at pH 12, which is an elongated globular molecule, suggesting that oleic acid inhibits the folding or collapse of the protein component of BAMLET to the globular form. Circular dichroism of BAMLET and apo-α-lactalbumin was performed and the results suggest that α-lactalbumin and BAMLET unfold in a continuum of increasing degree of unfolded states. Taken together, these results support a model in which BAMLET retains oleic acid by non-specific association in the core of partially unfolded protein, and represent a new type of lipoprotein structure.
Collapse
Affiliation(s)
- Emma M Rath
- Faculty of Pharmacy, University of Sydney, Sydney, New South Wales, 2006, Australia
| | | | | | | | | |
Collapse
|
34
|
Lopez S, Bermudez B, Montserrat-de la Paz S, Jaramillo S, Varela LM, Ortega-Gomez A, Abia R, Muriana FJG. Membrane composition and dynamics: a target of bioactive virgin olive oil constituents. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:1638-56. [PMID: 24440426 DOI: 10.1016/j.bbamem.2014.01.007] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 01/03/2014] [Accepted: 01/07/2014] [Indexed: 12/26/2022]
Abstract
The endogenous synthesis of lipids, which requires suitable dietary raw materials, is critical for the formation of membrane bilayers. In eukaryotic cells, phospholipids are the predominant membrane lipids and consist of hydrophobic acyl chains attached to a hydrophilic head group. The relative balance between saturated, monounsaturated, and polyunsaturated acyl chains is required for the organization and normal function of membranes. Virgin olive oil is the richest natural dietary source of the monounsaturated lipid oleic acid and is one of the key components of the healthy Mediterranean diet. Virgin olive oil also contains a unique constellation of many other lipophilic and amphipathic constituents whose health benefits are still being discovered. The focus of this review is the latest evidence regarding the impact of oleic acid and the minor constituents of virgin olive oil on the arrangement and behavior of lipid bilayers. We highlight the relevance of these interactions to the potential use of virgin olive oil in preserving the functional properties of membranes to maintain health and in modulating membrane functions that can be altered in several pathologies. This article is part of a Special Issue entitled: Membrane Structure and Function: Relevance in the Cell's Physiology, Pathology and Therapy.
Collapse
Affiliation(s)
- Sergio Lopez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41012 Seville, Spain
| | - Beatriz Bermudez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41012 Seville, Spain
| | | | - Sara Jaramillo
- Laboratory of Phytochemicals and Food Quality, Instituto de la Grasa, CSIC, 41014 Seville, Spain
| | - Lourdes M Varela
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41012 Seville, Spain
| | - Almudena Ortega-Gomez
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41012 Seville, Spain
| | - Rocio Abia
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41012 Seville, Spain
| | - Francisco J G Muriana
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, 41012 Seville, Spain.
| |
Collapse
|
35
|
Hoque M, Dave S, Gupta P, Saleemuddin M. Oleic acid may be the key contributor in the BAMLET-induced erythrocyte hemolysis and tumoricidal action. PLoS One 2013; 8:e68390. [PMID: 24039698 PMCID: PMC3770648 DOI: 10.1371/journal.pone.0068390] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 06/03/2013] [Indexed: 12/19/2022] Open
Abstract
A chance discovery of the tumoricidal action of a human milk fraction led to the characterization of the active component as oleic acid complex of the α-lactalbumin, which was given the acronym HAMLET. We report in this study that the oleic acid complex of bovine α-lactalbumin (BAMLET) is hemolytic to human erythrocytes as well as to those derived from some other mammals. Indirect immunofluorescence analysis suggested binding of BAMLET to erythrocytes prior to induction of hemolysis. Free OA was hemolytic albeit at higher concentrations, while sodium oleate caused hemolysis at far lower concentrations. Amiloride and BaCl2 offered protection against BAMLET-induced hemolysis suggesting the involvement of a cation leak channel in the process. BAMLET coupled to CNBr-activated Sepharose was not only hemolytic but also tumoricidal to Jurkat and MCF-7 cells in culture. The Sepharose-linked preparation was however not toxic to non-cancerous peritoneal macrophages and primary adipocytes. The tumoricidal action was studied using the MTT-assay while apoptosis induction measured by the annexin V-propidium iodide assay. Repeated incubation of the immobilized BAMLET with erythrocytes depleted oleic acid and decreased the hemolytic activity of the complex. Incubation of MCF-7 and Jurkat cells with OA, soluble or immobilized BAMLET resulted in increase in the uptake of Lyso Tracker Red and Nile red by the cells. The data presented support the contention that oleic acid plays the key role, both in BAMLET-induced hemolysis and tumoricidal action.
Collapse
Affiliation(s)
- Mehboob Hoque
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Sandeep Dave
- Institute of Microbial Technology (CSIR), Chandigarh, India
| | - Pawan Gupta
- Institute of Microbial Technology (CSIR), Chandigarh, India
| | | |
Collapse
|
36
|
Nemashkalova EL, Kazakov AS, Khasanova LM, Permyakov EA, Permyakov SE. Structural characterization of more potent alternatives to HAMLET, a tumoricidal complex of α-lactalbumin and oleic acid. Biochemistry 2013; 52:6286-99. [PMID: 23947814 DOI: 10.1021/bi400643s] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
HAMLET is a complex of human α-lactalbumin (hLA) with oleic acid (OA) that kills various tumor cells and strains of Streptococcus pneumoniae. More potent protein-OA complexes were previously reported for bovine α-lactalbumin (bLA) and β-lactoglobulin (bLG), and pike parvalbumin (pPA), and here we explore their structural features. The concentration dependencies of the tryptophan fluorescence of hLA, bLA, and bLG complexes with OA reveal their disintegration at protein concentrations below the micromolar level. Chemical cross-linking experiments provide evidence that association with OA shifts the distribution of oligomeric forms of hLA, bLA, bLG, and pPA toward higher-order oligomers. This effect is confirmed for bLA and bLG using the dynamic light scattering method, while pPA is shown to associate with OA vesicles. Like hLA binding, OA binding increases the affinity of bLG for small unilamellar dipalmitoylphosphatidylcholine vesicles, while pPA efficiently binds to the vesicles irrespective of OA binding. The association of OA with bLG and pPA increases their α-helix and cross-β-sheet content and resistance to enzymatic proteolysis, which is indicative of OA-induced protein structuring. The lack of excess heat sorption during melting of bLG and pPA in complex with OA and the presence of a cooperative thermal transition at the level of their secondary structure suggest that the OA-bound forms of bLG and pPA lack a fixed tertiary structure but exhibit a continuous thermal transition. Overall, despite marked differences, the HAMLET-like complexes that were studied exhibit a common feature: a tendency toward protein oligomerization. Because OA-induced oligomerization has been reported for other proteins, this phenomenon is inherent to many proteins.
Collapse
Affiliation(s)
- Ekaterina L Nemashkalova
- Institute for Biological Instrumentation of the Russian Academy of Sciences , Pushchino, Moscow region 142290, Russia
| | | | | | | | | |
Collapse
|