1
|
Ortiz-Hernandez GL, Sanchez-Hernandez ES, Ochoa PT, Casiano CA. The Emerging Roles of the Stress Epigenetic Reader LEDGF/p75 in Cancer Biology and Therapy Resistance: Mechanisms and Targeting Opportunities. Cancers (Basel) 2024; 16:3957. [PMID: 39682146 DOI: 10.3390/cancers16233957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
The lens epithelium derived growth factor of 75 kD (LEDGF/p75) is a transcription co-activator and epigenetic reader that has emerged as a stress oncoprotein in multiple human cancers. Growing evidence indicates that it promotes tumor cell survival against certain therapeutic drugs. The amino (N)-terminal region of LEDGF/p75 contains a PWWP domain that reads methylated histone marks, critical for recognizing transcriptionally active chromatin sites. Its carboxyl (C)-terminus has an integrase binding domain (IBD) that serves as the binding site for the HIV-1 integrase and multiple oncogenic transcription factors. Acting as hubs for protein-protein interactions, both domains facilitate the tethering of oncogenic transcription factors and regulators to active chromatin to regulate mRNA splicing, promote DNA repair, and enhance the expression of stress and cancer-related genes that contribute to tumor cell aggressiveness and chemoresistance. This review summarizes our current knowledge of the emerging roles of LEDGF/p75 in cancer biology and therapy resistance and discusses its potential as a novel oncotherapeutic target in combinatorial treatments.
Collapse
Affiliation(s)
- Greisha L Ortiz-Hernandez
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Evelyn S Sanchez-Hernandez
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Pedro T Ochoa
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Carlos A Casiano
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Department of Medicine, Division of Rheumatology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Cancer Center, Loma Linda University Health, Loma Linda, CA 92350, USA
| |
Collapse
|
2
|
Tran HT, Wan MLY, Ambite I, Cavalera M, Grossi M, Háček J, Esmaeili P, Carneiro ANBM, Chaudhuri A, Ahmadi S, Svanborg C. BAMLET administration via drinking water inhibits intestinal tumor development and promotes long-term health. Sci Rep 2024; 14:3838. [PMID: 38360830 PMCID: PMC10869698 DOI: 10.1038/s41598-024-54040-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/07/2024] [Indexed: 02/17/2024] Open
Abstract
Though new targeted therapies for colorectal cancer, which progresses from local intestinal tumors to metastatic disease, are being developed, tumor specificity remains an important problem, and side effects a major concern. Here, we show that the protein-fatty acid complex BAMLET (bovine alpha-lactalbumin made lethal to tumor cells) can act as a peroral treatment for colorectal cancer. ApcMin/+ mice, which carry mutations relevant to hereditary and sporadic human colorectal cancer, that received BAMLET in the drinking water showed long-term protection against tumor development and decreased expression of tumor growth-, migration-, metastasis- and angiogenesis-related genes. BAMLET treatment via drinking water inhibited the Wnt/β-catenin and PD-1 signaling pathways and prolonged survival without evidence of toxicity. Systemic disease in the lungs, livers, spleens, and kidneys, which accompanied tumor progression, was inhibited by BAMLET treatment. The metabolic response to BAMLET included carbohydrate and lipid metabolism, which were inhibited in tumor prone ApcMin/+ mice and weakly regulated in C57BL/6 mice, suggesting potential health benefits of peroral BAMLET administration in addition to the potent antitumor effects. Together, these findings suggest that BAMLET administration in the drinking water maintains antitumor pressure by removing emergent cancer cells and reprogramming gene expression in intestinal and extra-intestinal tissues.
Collapse
Affiliation(s)
- Hien Thi Tran
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Murphy Lam Yim Wan
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Ines Ambite
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Michele Cavalera
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Mario Grossi
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Jaromir Háček
- Department of Pathology and Molecular Medicine, Motol University Hospital, 2nd Faculty of Medicine, Charles University Praha, 150 06, Prague, Czech Republic
| | - Parisa Esmaeili
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - António N B M Carneiro
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Arunima Chaudhuri
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Shahram Ahmadi
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden
| | - Catharina Svanborg
- Division of Microbiology, Immunology and Glycobiology, Department of Laboratory Medicine, Faculty of Medicine, Lund University, Klinikgatan 28, 221 84, Lund, Sweden.
| |
Collapse
|
3
|
Xiao J, Ma J, Khan MZ, Alugongo GM, Chen T, Liu S, Li S, Cao Z. Unlocking the potential of milk whey protein components in colorectal cancer prevention and therapy. Crit Rev Food Sci Nutr 2023; 64:12961-12998. [PMID: 37846905 DOI: 10.1080/10408398.2023.2258970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Extensive research from large prospective cohort studies and meta-analytical investigations over recent decades have consistently indicated that dairy foods have protective effects, reducing the risk of colorectal cancer. Most of the literature has explored the potential role of milk minerals and vitamins in managing colorectal cancer. Yet, there is a paucity of a comprehensive summary of the anticancer attributes of milk protein components and their underlying mechanisms of action. Recent advancements have spotlighted the potential of whey proteins, including β-lactoglobulin, α-lactalbumin, serum albumin, and lactoferrin, as promising candidates for both the prevention and treatment of colorectal cancer. Notably, whey proteins have demonstrated a more pronounced capacity for suppressing carcinogen-induced tumors when compared to casein. Their strong binding affinity enables them to serve as effective carriers for small molecules or drugs targeting colon cancer therapy. Furthermore, numerous studies have underscored the anti-inflammatory and antioxidant prowess of whey proteins in cancer prevention. Additionally, whey proteins have been shown to trigger apoptosis, hinder tumor cell proliferation, and impede metastasis. This comprehensive review, therefore, not only substantiates the significance of incorporating whey protein components into a balanced daily diet but also underscores their potential in safeguarding against the onset and progression of colorectal cancer.
Collapse
Affiliation(s)
- Jianxin Xiao
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Key Laboratory of Low Carbon Culture and Safety Production in Cattle in Sichuan, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Jiaying Ma
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Muhammad Zahoor Khan
- Faculty of Veterinary and Animal Sciences, University of Agriculture Dera Ismail Khan, Khyber Pakhtunkhwa, Pakistan
| | - Gibson Maswayi Alugongo
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Tianyu Chen
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shuai Liu
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition and Feeding, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
4
|
Sanchez-Hernandez ES, Ochoa PT, Suzuki T, Ortiz-Hernandez GL, Unternaehrer JJ, Alkashgari HR, Diaz Osterman CJ, Martinez SR, Chen Z, Kremsky I, Wang C, Casiano CA. Glucocorticoid Receptor Regulates and Interacts with LEDGF/p75 to Promote Docetaxel Resistance in Prostate Cancer Cells. Cells 2023; 12:2046. [PMID: 37626856 PMCID: PMC10453226 DOI: 10.3390/cells12162046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Patients with advanced prostate cancer (PCa) invariably develop resistance to anti-androgen therapy and taxane-based chemotherapy. Glucocorticoid receptor (GR) has been implicated in PCa therapy resistance; however, the mechanisms underlying GR-mediated chemoresistance remain unclear. Lens epithelium-derived growth factor p75 (LEDGF/p75, also known as PSIP1 and DFS70) is a glucocorticoid-induced transcription co-activator implicated in cancer chemoresistance. We investigated the contribution of the GR-LEDGF/p75 axis to docetaxel (DTX)-resistance in PCa cells. GR silencing in DTX-sensitive and -resistant PCa cells decreased LEDGF/p75 expression, and GR upregulation in enzalutamide-resistant cells correlated with increased LEDGF/p75 expression. ChIP-sequencing revealed GR binding sites in the LEDGF/p75 promoter. STRING protein-protein interaction analysis indicated that GR and LEDGF/p75 belong to the same transcriptional network, and immunochemical studies demonstrated their co-immunoprecipitation and co-localization in DTX-resistant cells. The GR modulators exicorilant and relacorilant increased the sensitivity of chemoresistant PCa cells to DTX-induced cell death, and this effect was more pronounced upon LEDGF/p75 silencing. RNA-sequencing of DTX-resistant cells with GR or LEDGF/p75 knockdown revealed a transcriptomic overlap targeting signaling pathways associated with cell survival and proliferation, cancer, and therapy resistance. These studies implicate the GR-LEDGF/p75 axis in PCa therapy resistance and provide a pre-clinical rationale for developing novel therapeutic strategies for advanced PCa.
Collapse
Affiliation(s)
- Evelyn S. Sanchez-Hernandez
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
| | - Pedro T. Ochoa
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
| | - Tise Suzuki
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
| | - Greisha L. Ortiz-Hernandez
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
| | - Juli J. Unternaehrer
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
| | - Hossam R. Alkashgari
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Department of Physiology, College of Medicine, University of Jeddah, Jeddah 23890, Saudi Arabia
| | - Carlos J. Diaz Osterman
- Department of Basic Sciences, Ponce Health Sciences University, Ponce, PR 00716, USA; (C.J.D.O.); (S.R.M.)
| | - Shannalee R. Martinez
- Department of Basic Sciences, Ponce Health Sciences University, Ponce, PR 00716, USA; (C.J.D.O.); (S.R.M.)
| | - Zhong Chen
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Isaac Kremsky
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Charles Wang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Center for Genomics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Carlos A. Casiano
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (E.S.S.-H.); (T.S.); (G.L.O.-H.); (J.J.U.); (H.R.A.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (Z.C.); (I.K.); (C.W.)
- Rheumatology Division, Department of Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
5
|
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
|
6
|
The Comparative Role of BAMLET and 5-Fluorouracil in Colorectal Cancer Cells by Targeting WNT/& Beta; -Catenin Pathway. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2022. [DOI: 10.5812/ijcm-123140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background: Aberrant activation of the WNT/β-catenin signaling pathway is involved in various types of cancers, particularly colorectal cancer (CRC), which is a prevalent malignancy. Targeting the Wnt signaling pathway has gained a reputation as an attractive therapeutic strategy, mainly because of its potential for regulating cell proliferation, migration, differentiation, angiogenesis, and apoptosis. Objectives: The aim of the current research was to investigate the effects of 5-Fluorouracil (5-FU) and bovine alpha-lactalbumin made lethal to tumor cells (BAMLET), a complex of oleic acid with bovine α-lactalbumin protein, on colon cancer cells focusing on the Wnt signaling pathway. Methods: For this purpose, HT-29 and HCT116 cells were treated with 5-FU and BAMLET, and the expression levels of Wnt signaling-related proteins (β-catenin and E-cadherin) and VEGF as angiogenesis regulators were evaluated by quantitative real-time polymerase chain reaction (RT-qPCR) and Western Blot analysis. Results: Bovine alpha-lactalbumin made lethal to tumor cells (BAMLET) treatment down-regulated the expression of β-catenin and up-regulated the expression of E-cadherin significantly compared to the 5-FU (P < 0.0001). The reduced mRNA levels of VEGF in treated cells revealed the effectiveness of 5-FU and BAMLET on angiogenesis. Conclusions: Bovine alpha-lactalbumin made lethal to tumor cells (BAMLET) can be considered for targeting the Wnt signaling pathway and angiogenesis. It is amenable to further investigation in the development of CRC treatment.
Collapse
|
7
|
Ho JCS, Mir SA, Cavalera M, Esmaeili P, Tran TH, Yann ZC, Tran TH, Chaudhuri A, Bendt AK, Wenk MR, Svanborg C. Lipid bilayer composition as a determinant of cancer cell sensitivity to tumoricidal protein-lipid complexes. Biofactors 2022; 48:1145-1159. [PMID: 35388547 DOI: 10.1002/biof.1841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/24/2022] [Indexed: 11/10/2022]
Abstract
Complexes formed by the alpha1 N-terminal peptide of alpha-lactalbumin and oleic acid (alpha1-oleate) interact with lipid bilayers. Plasma membrane perturbations trigger tumor cell death but normal differentiated cells are more resistant, and their plasma membranes are less strongly affected. This study examined membrane lipid composition as a determinant of tumor cell reactivity. Bladder cancer tissue showed a higher abundance of unsaturated lipids enriched in phosphatidylcholine, PC (36:4) and PC (38:4), and sphingomyelin, SM (36:1) than healthy bladder tissue, where saturated lipids predominated and the lipid extracts from bladder cancer tissue inhibited the tumoricidal effect of the complex more effectively than healthy tissue extracts. Furthermore, unsaturated PC in solution inhibited tumor cell death, and the complex interacted with giant unilamellar vesicles formed by PC, confirming the affinity of alpha1-oleate for fluid membranes enriched in PC. Quartz Crystal Microbalance with dissipation monitoring (QCM-D) detected a preference of the complex for the liquid-disordered phase, suggesting that the insertion into PC-based membranes and the resulting membrane perturbations are influenced by membrane lipid saturation. The results suggest that the membrane lipid composition is functionally important and that specific unsaturated membrane lipids may serve as "recognition motifs" for broad-spectrum tumoricidal molecules such as alpha1-oleate.
Collapse
Affiliation(s)
- James C S Ho
- School of Materials Science & Engineering, Nanyang Technological University, Singapore, Singapore
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Sartaj Ahmad Mir
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michele Cavalera
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Parisa Esmaeili
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Tuan Hiep Tran
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Zandra Chew Yann
- School of Materials Science & Engineering, Nanyang Technological University, Singapore, Singapore
| | - Thi Hien Tran
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Arunima Chaudhuri
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Anne K Bendt
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Markus R Wenk
- Singapore Lipidomics Incubator (SLING), Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore, Singapore
| | - Catharina Svanborg
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
8
|
Comparative Analysis of the Protein Composition of Goat Milk from French Alpine, Nubian, and Creole Breeds and Holstein Friesian Cow Milk: Implications for Early Infant Nutrition. Animals (Basel) 2022; 12:ani12172236. [PMID: 36077959 PMCID: PMC9454708 DOI: 10.3390/ani12172236] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Goat’s milk is a food that contains proteins of value for nutrition. The protein profile in the milk of goat breeds is different from that of cow milk, with a lower relative abundance of allergenic proteins. In addition, regardless of the breed, goat milk has beta-casein of type A2 in a more significant proportion than cow milk, which impacts different bioactive peptides hydrolyzed in the milk of the species. Abstract Of the diversity of proteins and high digestibility, goat milk will be a food of significant value for infant nutrition. The genetic polymorphisms of milk proteins play an essential role in the different degrees of allergic reactions. This work aimed to identify the proteins and peptides in the composition of goat milk and compare them to those in cow’s milk. The work was performed with goats French Alpine, Nubian, and Creole breeds and Holstein Friesian milking cows at the Universidad Autónoma de Querétaro, Amazcala. We investigated the relative abundance of goat and cow milk protein fractions by SDS-PAGE resolution and the densitometric analysis of gels. The protein alfa-casein was (17.67 ± 0.46) for Creole, (19.18 ± 0.88) French Alpine, (17.35 ± 0.49) Nubian, and (35.92 ± 1.96) Holstein cows. The relative abundance obtained from alfa-casein was statistically different between goats and cows, and this protein was vital because it is a protein related to allergies. On the other hand, the amino acid in position 67 of the beta-casein from three goat breeds is a Proline, so it is assumed that the beta-casein variant of goat milk is an A2-type. The latter has excellent relevance for infant nutrition and differs from cow milk.
Collapse
|
9
|
Tarapata J, Maciejczyk M, Zulewska J. Microfiltration of buttermilk: partitioning of proteins and modelling using a resistance-in-series model. Int Dairy J 2022. [DOI: 10.1016/j.idairyj.2022.105445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
10
|
Pei W, Cai L, Gong X, Zhang L, Zhang J, Zhu P, Jiang H, Wang C, Wang S, Chen J. Drug-loaded oleic-acid grafted mesoporous silica nanoparticles conjugated with α-lactalbumin resembling BAMLET-like anticancer agent with improved biocompatibility and therapeutic efficacy. Mater Today Bio 2022; 15:100272. [PMID: 35607417 PMCID: PMC9123267 DOI: 10.1016/j.mtbio.2022.100272] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 11/21/2022] Open
Abstract
Despite its prominent therapeutic efficacy, chemotherapy has raised serious concerns due to the severe adverse effects and multidrug resistance evoked, which propels the search for safe and green therapeutic agents. BAMLET (bovine α-lactalbumin made lethal against tumor cell) is a well-known protein-based anticancer agent of selective tumoricidal activity. Here, we prepared oleic acid-modified mesoporous silica nanoparticles (OA-MSNs) conjugated with bovine α-lactalbumin, a lipoprotein complex resembling BAMLET formed on the surface of MSNs (MSN-BAMLET) to load the anticancer drug of docetaxel (DTX). Compared to that of OA-MSNs/DTX, the obtained MSN-BAMLET/DTX with a sustained and pH-responsive drug release behaviors exhibited good biocompatibility and enhanced cytotoxic effect against cancer cells. Moreover, the presence of lipoprotein complex in MSN-BAMLET contributed to the improved dispersion of the composite in solution and the inhibitory effect on the migration of cancer cells. Furthermore, the adsorption profiles of protein corona on the obtained nanoparticles were analyzed. It was found that the marked low amount and abundance of plasma proteins were adsorbed on the α-lactalbumin coated siliceous composite demonstrated its long circulation property. Finally, in vivo study showed that MSN-BAMLET/DTX contributed to the effective cancer ablation and the prolonged survival. Therefore, the constructed MSN-BAMLET of the mesoregular structure and peculiar tumoricidal effect provides a manipulable nanoplatform as drug nanocarrier for therapeutic applications.
Collapse
Affiliation(s)
- Wei Pei
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Ling Cai
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Xing Gong
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Li Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Jiarong Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Ping Zhu
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Huijun Jiang
- School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China
| | - Chao Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Shoulin Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
| | - Jin Chen
- Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 211166, Nanjing, China
- Jiangsu Province Engineering Research Center of Antibody Drug, Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, 211166, China
| |
Collapse
|
11
|
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
|
12
|
Chiou JT, Shi YJ, Lee YC, Wang LJ, Chen YJ, Chang LS. Carboxyl group-modified α-lactalbumin induces TNF-α-mediated apoptosis in leukemia and breast cancer cells through the NOX4/p38 MAPK/PP2A axis. Int J Biol Macromol 2021; 187:513-527. [PMID: 34310992 DOI: 10.1016/j.ijbiomac.2021.07.133] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/30/2022]
Abstract
To clarify the mechanism of semicarbazide-modified α-lactalbumin (SEM-LA)-mediated cytotoxicity, we investigated its effect on human U937 leukemia cells and MCF-7 breast cancer cells in the current study. SEM-LA induced apoptosis in U937 cells, which showed increased NOX4 expression, procaspase-8 degradation, and t-Bid production. FADD depletion inhibited SEM-LA-elicited caspase-8 activation, t-Bid production, and cell death, indicating that SEM-LA activated death receptor-mediated apoptosis in U937 cells. SEM-LA stimulated Ca2+-mediated Akt activation, which in turn increased Sp1- and p300-mediated NOX4 transcription. The upregulation of NOX4 expression promoted ROS-mediated p38 MAPK phosphorylation, leading to protein phosphatase 2A (PP2A)-regulated tristetraprolin (TTP) degradation. Remarkably, TTP downregulation increased the stability of TNF-α mRNA, resulting in the upregulation of TNF-α protein expression. Abolishment of Ca2+-NOX4-ROS axis-mediated p38 MAPK activation attenuated SEM-LA-induced TNF-α upregulation and protected U937 cells from SEM-LA-mediated cytotoxicity. The restoration of TTP expression alleviated the effect of TNF-α upregulation and cell death induced by SEM-LA. Altogether, the data in this study demonstrate that SEM-LA activates TNF-α-mediated apoptosis in U937 cells through the NOX4/p38 MAPK/PP2A axis. We think that a similar pathway can also explain the death of MCF-7 human breast cancer cells after SEM-LA treatment.
Collapse
Affiliation(s)
- Jing-Ting Chiou
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Yi-Jun Shi
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Yuan-Chin Lee
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Liang-Jun Wang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Ying-Jung Chen
- Department of Fragrance and Cosmetic Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
13
|
Leischner C, Egert S, Burkard M, Venturelli S. Potential Protective Protein Components of Cow's Milk against Certain Tumor Entities. Nutrients 2021; 13:1974. [PMID: 34201342 PMCID: PMC8228601 DOI: 10.3390/nu13061974] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 12/16/2022] Open
Abstract
Milk and dairy products, especially from cow's milk, play a major role in the daily human diet. It is therefore hardly surprising that the subject of milk is being extensively researched and that many effects of individual milk components have been characterized as a result. With the wealth of results available today, the influence of milk on the development of various types of cancer and, in particular, its often protective effects have been shown both in vitro and in vivo and in the evaluation of large-scale cohort and case-control studies. Various caseins, diverse whey proteins such as α-lactalbumin (α-LA), bovine α-lactalbumin made lethal to tumor cells (BAMLET), β-lactoglobulin (β-LG), or bovine serum albumin (BSA), and numerous milk fat components, such as conjugated linoleic acid (CLA), milk fat globule membrane (MFGM), or butyrate, as well as calcium and other protein components such as lactoferrin (Lf), lactoferricin (Lfcin), and casomorphines, show antitumor or cytotoxic effects on cells from different tumor entities. With regard to a balanced and health-promoting diet, milk consumption plays a major role in a global context. This work provides an overview of what is known about the antitumoral properties of proteins derived from cow's milk and their modes of action.
Collapse
Affiliation(s)
- Christian Leischner
- Institute of Nutritional Sciences 140, Nutritional Biochemistry 140c, University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany;
| | - Sarah Egert
- Institute of Nutritional Medicine, Nutritional Science/Dietetics 180c, University of Hohenheim, Fruwirthstr. 12, 70599 Stuttgart, Germany;
| | - Markus Burkard
- Institute of Nutritional Sciences 140, Nutritional Biochemistry 140c, University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany;
| | - Sascha Venturelli
- Institute of Nutritional Sciences 140, Nutritional Biochemistry 140c, University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany;
- Department of Vegetative and Clinical Physiology, Institute of Physiology, University Hospital Tuebingen, Wilhelmstr. 56, 72074 Tuebingen, Germany
| |
Collapse
|
14
|
Rocha-Mendoza D, Kosmerl E, Krentz A, Zhang L, Badiger S, Miyagusuku-Cruzado G, Mayta-Apaza A, Giusti M, Jiménez-Flores R, García-Cano I. Invited review: Acid whey trends and health benefits. J Dairy Sci 2020; 104:1262-1275. [PMID: 33358165 DOI: 10.3168/jds.2020-19038] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022]
Abstract
In recent years, acid whey production has increased due to a growing demand for Greek yogurt and acid-coagulated cheeses. Acid whey is a dairy by-product for which the industry has long struggled to find a sustainable application. Bulk amounts of acid whey associated with the dairy industry have led to increasing research on ways to valorize it. Industry players are finding ways to use acid whey on-site with ultrafiltration techniques and biodigesters, to reduce transportation costs and provide energy for the facility. Academia has sought to further investigate practical uses and benefits of this by-product. Although modern research has shown many other possible applications for acid whey, no comprehensive review yet exists about its composition, utilization, and health benefits. In this review, the industrial trends, the applications and uses, and the potential health benefits associated with the consumption of acid whey are discussed. The proximal composition of acid whey is discussed in depth. In addition, the potential applications of acid whey, such as its use as a starting material in the production of fermented beverages, as growth medium for cultivation of lactic acid bacteria in replacement of commercial media, and as a substrate for the isolation of lactose and minerals, are reviewed. Finally, the potential health benefits of the major protein constituents of acid whey, bioactive phospholipids, and organic acids such as lactic acid are described. Acid whey has promising applications related to potential health benefits, ranging from antibacterial effects to cognitive development for babies to human gut health.
Collapse
Affiliation(s)
- Diana Rocha-Mendoza
- Department of Food Science and Technology, The Ohio State University, Columbus 43210
| | - Erica Kosmerl
- Department of Food Science and Technology, The Ohio State University, Columbus 43210
| | - Abigail Krentz
- Department of Food Science and Technology, The Ohio State University, Columbus 43210
| | - Lin Zhang
- Department of Food Science and Technology, The Ohio State University, Columbus 43210
| | - Shivani Badiger
- Department of Food Science and Technology, The Ohio State University, Columbus 43210
| | | | - Alba Mayta-Apaza
- Department of Food Science and Technology, The Ohio State University, Columbus 43210
| | - Monica Giusti
- Department of Food Science and Technology, The Ohio State University, Columbus 43210
| | - Rafael Jiménez-Flores
- Department of Food Science and Technology, The Ohio State University, Columbus 43210.
| | - Israel García-Cano
- Department of Food Science and Technology, The Ohio State University, Columbus 43210.
| |
Collapse
|
15
|
Kalouta K, Stie MB, Janfelt C, Chronakis IS, Jacobsen J, Mørck Nielsen H, Foderà V. Electrospun α-Lactalbumin Nanofibers for Site-Specific and Fast-Onset Delivery of Nicotine in the Oral Cavity: An In Vitro, Ex Vivo, and Tissue Spatial Distribution Study. Mol Pharm 2020; 17:4189-4200. [PMID: 32885978 DOI: 10.1021/acs.molpharmaceut.0c00642] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Nicotine replacement therapy (NRT) formulations for oromucosal administration induce a delayed rise in nicotine blood levels as opposed to the immediate nicotine increase obtained from cigarette smoking, this being a shortcoming of the therapy. Here, we demonstrate that α-lactalbumin/polyethylene oxide (ALA/PEO) electrospun nanofibers constitute an efficient oromucosal delivery system for fast-onset nicotine delivery of high relevance for acute dosing NRT applications. In vitro, nicotine-loaded nanofibers showed fast disintegration in water, with a weight loss up to 40% within minutes, and a faster nicotine release (26.1 ± 4.6% after 1 min of incubation) of the loaded nicotine compared to two relevant marketed NRT formulations with a comparable nicotine dose (i.e., 7.9 ± 5.1 and 2.2 ± 0.3% nicotine was released from a lozenge and a sublingual tablet, respectively). Model-fitting of the release data indicated that the release mechanism of nicotine from the hydrophilic nanofibers was possibly governed by more than one type of release phenomena. Remarkably, ex vivo studies using porcine buccal mucosa demonstrated a more efficient permeation of the nicotine released from the nanofibers [flux of 1.06 ± 0.22 nmol/(cm2·min)] compared to when dosing even a ten-fold concentrated nicotine solution [flux of 0.17 ± 0.14 nmol/(cm2·min)]. Moreover, matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI MS) imaging of ex vivo porcine buccal mucosa exposed to nicotine-loaded nanofibers clearly revealed higher amounts of nicotine throughout the epithelium, as well as in the lamina propria and submucosa of the tissue. Our findings suggest that nicotine-loaded ALA/PEO nanofibers have potential as a mucosal, fast-releasing, and biocompatible delivery system for nicotine, which can overcome the limitations of the currently marketed NRTs.
Collapse
Affiliation(s)
- Kleopatra Kalouta
- Department of Pharmacy, University of Copenhagen, 2 Universitetsparken, 2100 Copenhagen, Denmark.,Center for Biopharmaceuticals and Biobarriers in Drug Delivery, University of Copenhagen, 2 Universitetsparken, 2100 Copenhagen, Denmark
| | - Mai Bay Stie
- Department of Pharmacy, University of Copenhagen, 2 Universitetsparken, 2100 Copenhagen, Denmark.,Center for Biopharmaceuticals and Biobarriers in Drug Delivery, University of Copenhagen, 2 Universitetsparken, 2100 Copenhagen, Denmark
| | - Christian Janfelt
- Department of Pharmacy, University of Copenhagen, 2 Universitetsparken, 2100 Copenhagen, Denmark
| | - Ioannis S Chronakis
- DTU Food, Technical University of Denmark, Kemitorvet, B202, 2800 Kgs. Lyngby, Denmark
| | - Jette Jacobsen
- Department of Pharmacy, University of Copenhagen, 2 Universitetsparken, 2100 Copenhagen, Denmark
| | - Hanne Mørck Nielsen
- Department of Pharmacy, University of Copenhagen, 2 Universitetsparken, 2100 Copenhagen, Denmark.,Center for Biopharmaceuticals and Biobarriers in Drug Delivery, University of Copenhagen, 2 Universitetsparken, 2100 Copenhagen, Denmark
| | - Vito Foderà
- Department of Pharmacy, University of Copenhagen, 2 Universitetsparken, 2100 Copenhagen, Denmark.,Center for Biopharmaceuticals and Biobarriers in Drug Delivery, University of Copenhagen, 2 Universitetsparken, 2100 Copenhagen, Denmark
| |
Collapse
|
16
|
Liu J, Chen WM, Shao YH, Liu YP, Tu ZC. Improved antitumor activity and IgE/IgG-binding ability of α-Lactalbumin/β-lactoglobulin induced by ultrasonication prior to binding with oleic acid. J Food Biochem 2020; 44:e13502. [PMID: 33025647 DOI: 10.1111/jfbc.13502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/11/2020] [Accepted: 09/01/2020] [Indexed: 12/29/2022]
Abstract
Bovine α-lactalbumin (α-La)/β-lactoglobulin (β-Lg) was pretreated through ultrasonic treatment and subsequently binding with oleic acid (OA) by heat treatment. And, the antitumor activity, IgE/IgG-binding ability, and structural modifications were investigated. After α-La/β-Lg were treated by ultrasonic prior to binding with OA, the treated α-La/β-Lg showed high antitumor activity and IgE/IgG-binding ability, and significantly affected the structural modifications, which reflected by the reduction in α-helix content, the increase of molecular weight, intrinsic fluorescence intensity, and surface hydrophobicity. Molecular docking studies indicated that OA bound to α-La/β-Lg by hydrogen bonds and hydrophobic interaction. Therefore, ultrasonic prior to binding with OA could improve antitumor activity and IgE/IgG-binding ability of α-La/β-Lg as a result of structural modifications. And, ultrasonic prior to binding with fatty acid processing of milk products alone may increase the antitumor activity, this change may enhance the risk of an allergenic reaction in milk allergy patients to some extent. PRACTICAL APPLICATIONS: Fatty acids, natural ligands associated with the bovine milk proteins, and milk protein-fatty acid complex has a variety of functional applications in the food industry. This study revealed that antitumor activity, IgE/IgG-binding ability, and structural modifications of α-La/β-Lg induced by ultrasonic prior to binding with oleic acid. It will be beneficial to understand the mechanism of the functional changes of protein. Ultrasonic prior to binding with oleic acid will be more likely to develop a practical technology to improve the functional characteristics of milk protein and design the optimal nutritional performance of milk food.
Collapse
Affiliation(s)
- Jun Liu
- National Research and Development center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, China.,Engineering Research Center of Freshwater Fish High-value Utilization of Jiangxi Province, Nanchang, China
| | - Wen-Mei Chen
- National Research and Development center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, China.,Engineering Research Center of Freshwater Fish High-value Utilization of Jiangxi Province, Nanchang, China
| | - Yan-Hong Shao
- National Research and Development center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, China.,Engineering Research Center of Freshwater Fish High-value Utilization of Jiangxi Province, Nanchang, China
| | - Ying-Ping Liu
- National Research and Development center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, China
| | - Zong-Cai Tu
- National Research and Development center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, China.,Engineering Research Center of Freshwater Fish High-value Utilization of Jiangxi Province, Nanchang, China.,State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
17
|
Nagakannan P, Tabeshmehr P, Eftekharpour E. Oxidative damage of lysosomes in regulated cell death systems: Pathophysiology and pharmacologic interventions. Free Radic Biol Med 2020; 157:94-127. [PMID: 32259579 DOI: 10.1016/j.freeradbiomed.2020.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/16/2022]
Abstract
Lysosomes are small specialized organelles containing a variety of different hydrolase enzymes that are responsible for degradation of all macromolecules, entering the cells through the endosomal system or originated from the internal sources. This allows for transport and recycling of nutrients and internalization of surface proteins for antigen presentation as well as maintaining cellular homeostasis. Lysosomes are also important storage compartments for metal ions and nutrients. The integrity of lysosomal membrane is central to maintaining their normal function, but like other cellular membranes, lysosomal membrane is subject to damage mediated by reactive oxygen species. This results in spillage of lysosomal enzymes into the cytoplasm, leading to proteolytic damage to cellular systems and organelles. Several forms of lysosomal dependent cell death have been identified in diseases. Examination of these events are important for finding treatment strategies relevant to cancer or neurodegenerative diseases as well as autoimmune deficiencies. In this review, we have examined the current literature on involvement of lysosomes in induction of programed cell death and have provided an extensive list of therapeutic approaches that can modulate cell death. Exploitation of these mechanisms can lead to novel therapies for cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Pandian Nagakannan
- Regenerative Medicine Program and Spinal Cord Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Parisa Tabeshmehr
- Regenerative Medicine Program and Spinal Cord Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Eftekhar Eftekharpour
- Regenerative Medicine Program and Spinal Cord Research Centre, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
18
|
α-Lactalbumin, Amazing Calcium-Binding Protein. Biomolecules 2020; 10:biom10091210. [PMID: 32825311 PMCID: PMC7565966 DOI: 10.3390/biom10091210] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [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
|
19
|
The novel therapeutic potential of bovine α-lactalbumin made lethal to tumour cells (BALMET) and oleic acid in oral squamous cell carcinoma (OSCC). Eur J Cancer Prev 2020; 30:178-187. [PMID: 32694279 DOI: 10.1097/cej.0000000000000617] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Since the serendipitous discovery of bovine α-lactalbumin made lethal to tumour cells (BAMLET)/human α-lactalbumin made lethal to tumour cells there has been an increased interest in the ability of the two components, oleic acid and α-lactalbumin, to form anti-cancer complexes. Here we have investigated the in-vitro efficacy of the BAMLET complex in killing oral cancer (OC) cells, determined the active component of the complex and investigated possible biological mechanisms. MATERIALS AND METHODS Two OC cell lines (±p53 mutation) and one dysplastic cell line were used as a model of progressive oral carcinogenesis. We performed cell viability assays with increasing BAMLET concentrations to determine the cytotoxic potential of the complex. We further analysed the individual components to determine their respective cytotoxicities. siRNA knockdown of p53 was used to determine its functional role in mediating sensitivity to BAMLET. Cell death mechanisms were investigated by flow cytometry, confocal microscopy and the lactate dehydrogenase assay. RESULTS Our results show that BAMLET is cytotoxic to the OC and dysplastic cell lines in a time and dose-dependent manner. The cytotoxic component was found to be oleic acid, which, can induce cytotoxicity even when not in complex. Our results indicate that the mechanism of cytotoxicity occurs through multiple simultaneous events including cell cycle arrest, autophagy like processes with a minor involvement of necrosis. CONCLUSION Deciphering the mechanism of cytotoxicity will aid treatment modalities for OC. This study highlights the potential of BAMLET as a novel therapeutic strategy in oral dysplastic and cancerous cells.
Collapse
|
20
|
Sabha BH, Masood A, Alanazi IO, Alfadda AA, Almehdar HA, Benabdelkamel H, Redwan EM. Comparative Analysis of Milk Fat Globular Membrane (MFGM) Proteome between Saudi Arabia Camelus dromedary Safra and Wadha Breeds. Molecules 2020; 25:E2146. [PMID: 32375319 PMCID: PMC7249027 DOI: 10.3390/molecules25092146] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 12/17/2022] Open
Abstract
Camel milk is traditionally known to have medicinal properties and many potential health benefits. Natural milk contains many soluble proteins and nanoparticles, such as a milk fat globule membrane (MFGM), a three-layered membrane covering of milk fat globule mainly composed of proteins and lipids, which plays an important role in human health. MFGM proteins account for 1%-4% of total milk proteins, and their nutritive value and distribution depends on the different breeds. The differential composition of these membrane proteins among different camel breeds has not been explored. The current study, therefore, aimed to quantitatively analyze and compare the MFGM proteome between the milk produced by the two most common Saudi camel breeds, Camelus dromedarius: Safra and Wadha. Two-dimensional difference in gel electrophoresis (2D-DIGE) and mass spectrometry analysis revealed a total of 44 MFGM proteins that were identified with a significant difference in abundance (p ≤ 0.05; fold change ≥ 1.5) between the two breeds. Thirty-one proteins were up-regulated and 13 proteins were down-regulated in the Safra breed compared to the Wadha breed. The proteins identified with an increased abundance included α-lactalbumin, lactadherin, and annexin a8, whereas the down-regulated proteins included butyrophilin subfamily 1 member a1, lactotransferrin, and vinculin. The differentially abundant proteins were analyzed by the UNIPROT system and gene ontology (GO) to reveal their associations with known biological functions and pathways. Enzyme-linked immunosorbent assay (ELISA) confirmed the 2D-DIGE findings of butyrophilin (BTN) and α-lactalbumin (α-LA) levels obtained from Safra and Wadha breeds.
Collapse
Affiliation(s)
- Bassam H. Sabha
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (B.H.S.); (H.A.A.)
| | - Afshan Masood
- Proteomics Unit, Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia; (A.M.); (A.A.A.)
| | - Ibrahim O. Alanazi
- The National Center for Genomic Technology (NCGT), Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), PO Box 6086, Riyadh 11461, Saudi Arabia;
| | - Assim A. Alfadda
- Proteomics Unit, Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia; (A.M.); (A.A.A.)
| | - Hussein A. Almehdar
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (B.H.S.); (H.A.A.)
| | - Hicham Benabdelkamel
- Proteomics Unit, Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia; (A.M.); (A.A.A.)
| | - Elrashdy M. Redwan
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (B.H.S.); (H.A.A.)
| |
Collapse
|
21
|
Ortiz-Hernandez GL, Sanchez-Hernandez ES, Casiano CA. Twenty years of research on the DFS70/LEDGF autoantibody-autoantigen system: many lessons learned but still many questions. AUTOIMMUNITY HIGHLIGHTS 2020; 11:3. [PMID: 32127038 PMCID: PMC7065333 DOI: 10.1186/s13317-020-0126-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/14/2020] [Indexed: 12/24/2022]
Abstract
The discovery and initial characterization 20 years ago of antinuclear autoantibodies (ANAs) presenting a dense fine speckled (DFS) nuclear pattern with strong staining of mitotic chromosomes, detected by indirect immunofluorescence assay in HEp-2 cells (HEp-2 IIFA test), has transformed our view on ANAs. Traditionally, ANAs have been considered as reporters of abnormal immunological events associated with the onset and progression of systemic autoimmune rheumatic diseases (SARD), also called ANA-associated rheumatic diseases (AARD), as well as clinical biomarkers for the differential diagnosis of these diseases. However, based on our current knowledge, it is not apparent that autoantibodies presenting the DFS IIF pattern fall into these categories. These antibodies invariably target a chromatin-associated protein designated as dense fine speckled protein of 70 kD (DFS70), also known as lens epithelium-derived growth factor protein of 75 kD (LEDGF/p75) and PC4 and SFRS1 Interacting protein 1 (PSIP1). This multi-functional protein, hereafter referred to as DFS70/LEDGF, plays important roles in the formation of transcription complexes in active chromatin, transcriptional activation of specific genes, regulation of mRNA splicing, DNA repair, and cellular survival against stress. Due to its multiple functions, it has emerged as a key protein contributing to several human pathologies, including acquired immunodeficiency syndrome (AIDS), leukemia, cancer, ocular diseases, and Rett syndrome. Unlike other ANAs, "monospecific" anti-DFS70/LEDGF autoantibodies (only detectable ANA in serum) are not associated with SARD and have been detected in healthy individuals and some patients with non-SARD inflammatory conditions. These observations have led to the hypotheses that these antibodies could be considered as negative biomarkers of SARD and might even play a protective or beneficial role. In spite of 20 years of research on this autoantibody-autoantigen system, its biological and clinical significance still remains enigmatic. Here we review the current state of knowledge of this system, focusing on the lessons learned and posing emerging questions that await further scrutiny as we continue our quest to unravel its significance and potential clinical and therapeutic utility.
Collapse
Affiliation(s)
- Greisha L Ortiz-Hernandez
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, USA
| | - Evelyn S Sanchez-Hernandez
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, USA
| | - Carlos A Casiano
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA. .,Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, USA. .,Department of Medicine/Division of Rheumatology, Loma Linda University School of Medicine, Loma Linda, USA.
| |
Collapse
|
22
|
Haller N, Kulozik U. Continuous centrifugal separation of selectively precipitated α-lactalbumin. Int Dairy J 2020. [DOI: 10.1016/j.idairyj.2019.104566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
23
|
Hämälistö S, Stahl JL, Favaro E, Yang Q, Liu B, Christoffersen L, Loos B, Guasch Boldú C, Joyce JA, Reinheckel T, Barisic M, Jäättelä M. Spatially and temporally defined lysosomal leakage facilitates mitotic chromosome segregation. Nat Commun 2020; 11:229. [PMID: 31932607 PMCID: PMC6957743 DOI: 10.1038/s41467-019-14009-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022] Open
Abstract
Lysosomes are membrane-surrounded cytoplasmic organelles filled with a powerful cocktail of hydrolases. Besides degrading cellular constituents inside the lysosomal lumen, lysosomal hydrolases promote tissue remodeling when delivered to the extracellular space and cell death when released to the cytosol. Here, we show that spatially and temporally controlled lysosomal leakage contributes to the accurate chromosome segregation in normal mammalian cell division. One or more chromatin-proximal lysosomes leak in the majority of prometaphases, after which active cathepsin B (CTSB) localizes to the metaphase chromatin and cleaves a small subset of histone H3. Stabilization of lysosomal membranes or inhibition of CTSB activity during mitotic entry results in a significant increase in telomere-related chromosome segregation defects, whereas cells and tissues lacking CTSB and cells expressing CTSB-resistant histone H3 accumulate micronuclei and other nuclear defects. These data suggest that lysosomal leakage and chromatin-associated CTSB contribute to proper chromosome segregation and maintenance of genomic integrity. Lysosomes are intracellular organelles containing degradative enzymes, and leakage of lysosomal contents into the cell is thought to trigger cell death. Here, the authors report that leaky lysosomes may facilitate chromosome separation during cell division.
Collapse
Affiliation(s)
- Saara Hämälistö
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Jonathan Lucien Stahl
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Elena Favaro
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Qing Yang
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Bin Liu
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Line Christoffersen
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, 7600, Stellenbosch, South Africa
| | - Claudia Guasch Boldú
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Johanna A Joyce
- Ludwig Institute for Cancer Research, University of Lausanne, 1005, Lausanne, Switzerland
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Medical Faculty, University of Freiburg, 79104, Freiburg, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, partner site Freiburg, 79106, Freiburg, Germany
| | - Marin Barisic
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark.,Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Marja Jäättelä
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, 2100, Copenhagen, Denmark. .,Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
24
|
Abstract
Being originally discovered as cellular recycling bins, lysosomes are today recognized as versatile signaling organelles that control a wide range of cellular functions that are essential not only for the well-being of normal cells but also for malignant transformation and cancer progression. In addition to their core functions in waste disposal and recycling of macromolecules and energy, lysosomes serve as an indispensable support system for malignant phenotype by promoting cell growth, cytoprotective autophagy, drug resistance, pH homeostasis, invasion, metastasis, and genomic integrity. On the other hand, malignant transformation reduces the stability of lysosomal membranes rendering cancer cells sensitive to lysosome-dependent cell death. Notably, many clinically approved cationic amphiphilic drugs widely used for the treatment of other diseases accumulate in lysosomes, interfere with their cancer-promoting and cancer-supporting functions and destabilize their membranes thereby opening intriguing possibilities for cancer therapy. Here, we review the emerging evidence that supports the supplementation of current cancer therapies with lysosome-targeting cationic amphiphilic drugs.
Collapse
|
25
|
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: 0.7] [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
|
26
|
Nadeem A, Ho JCS, Tran TH, Paul S, Granqvist V, Despretz N, Svanborg C. Beta-sheet-specific interactions with heat shock proteins define a mechanism of delayed tumor cell death in response to HAMLET. J Mol Biol 2019; 431:2612-2627. [PMID: 31082436 DOI: 10.1016/j.jmb.2019.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 05/02/2019] [Accepted: 05/04/2019] [Indexed: 02/07/2023]
Abstract
As chaperones, heat shock proteins (HSPs) protect host cells against misfolded proteins that constitute a by-product of protein synthesis. Certain HSPs are also expressed on the surface of tumor cells, possibly to scavenge extracellular unfolded protein ligands and prevent them from becoming cytotoxic. HAMLET-a complex of partially unfolded alpha-lactalbumin and oleic acid-is relying on its N-terminal alpha-helical domain to perturb tumor cell membranes, and the cells die as a consequence of this interaction. Here we show that in parallel, cell surface HSPs bind the beta-sheet domain of alpha-lactalbumin and activate a temporarily protective loop, involving vesicular uptake and lysosomal accumulation. Later, HAMLET destroys lysosomal membrane integrity, and HAMLET release kills the remaining tumor cells. HSPs were identified as HAMLET targets in a proteomic screen and Hsp70-specific antibodies or shRNAs inhibited HAMLET uptake by tumor cells, which showed increased Hsp70 surface expression compared to differentiated cells. The results suggest that HAMLET engages tumor cells by two parallel recognition mechanisms, defined by alpha-helical- or beta-sheet domains of alpha-lactalbumin and resulting in an immediate death response, or a delay due to transient accumulation of the complex in the lysosomes. This dual response pattern was conserved among tumor cells but not seen in normal, differentiated cells. By two different mechanisms, HAMLET thus achieves a remarkably efficient elimination of tumor cells.
Collapse
Affiliation(s)
- Aftab Nadeem
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden
| | - James C S Ho
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden; Centre for Biomimetic Sensor Science, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Drive, 637553, Singapore
| | - Tuan Hiep Tran
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden
| | - Sanchari Paul
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden
| | - Victoria Granqvist
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden
| | - Nadege Despretz
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden
| | - Catharina Svanborg
- Department of Microbiology, Immunology and Glycobiology (MIG), Institute of Laboratory Medicine, Lund University, S-223 62 Lund, Sweden.
| |
Collapse
|
27
|
Whey protein in cancer therapy: A narrative review. Pharmacol Res 2019; 144:245-256. [PMID: 31005617 DOI: 10.1016/j.phrs.2019.04.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 02/07/2023]
Abstract
Cancer remains a public health challenge in the identification and development of ideal pharmacological therapies and dietary strategies. The use of whey protein as a dietary strategy is widespread in the field of oncology. The two types of whey protein, sweet or acid, result from several processing techniques and possess distinct protein subfraction compositions. Mechanistically, whey protein subfractions have specific anti-cancer effects. Alpha-lactalbumin, human α-lactalbumin made lethal to tumor cell, bovine α-lactalbumin made lethal to tumor cell, bovine serum albumin, and lactoferrin are whey protein subfractions with potential to hinder tumor pathways. Such effects, however, are principally supported by studies performed in vitro and/or in vivo. In clinical practice, whey protein intake-induced anti-cancer effects are indiscernible. However, whey protein supplementation represents a practical, feasible, and cost-effective approach to mitigate cancer cachexia syndrome. The usefulness of whey protein is evidenced by a greater leucine content and the potential to modulate IGF-1 concentrations, representing important factors towards musculoskeletal hypertrophy. Further clinical trials are warranted and needed to establish the effects of whey protein supplementation as an adjuvant to cancer therapy.
Collapse
|
28
|
|
29
|
Dopierała K, Krajewska M, Prochaska K. Binding of α-lactalbumin to oleic acid monolayer and its relevance to formation of HAMLET-like complexes. Int Dairy J 2019. [DOI: 10.1016/j.idairyj.2018.08.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
30
|
Hsp70 interactions with membrane lipids regulate cellular functions in health and disease. Prog Lipid Res 2019; 74:18-30. [PMID: 30710597 DOI: 10.1016/j.plipres.2019.01.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/18/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023]
Abstract
Beyond guarding the cellular proteome the major stress inducible heat shock protein Hsp70 has been shown to interact with lipids. Non-cytosolic Hsp70 stabilizes membranes during stress challenges and, in pathophysiological states, facilitates endocytosis, counteracts apoptotic mechanisms, sustains survival pathways or represents a signal that can be recognized by the immune system. Disease-coupled lipid-associated functions of Hsp70 may be targeted via distinct subcellular localizations of Hsp70 itself or its specific interacting lipids. With a special focus on interacting lipids, here we discuss localization-dependent roles of the membrane-bound Hsp70 in the context of its therapeutic potential, particularly in cancer and neurodegenerative diseases.
Collapse
|
31
|
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]
|
32
|
Rath EM, Cheng YY, Pinese M, Sarun KH, Hudson AL, Weir C, Wang YD, Håkansson AP, Howell VM, Liu GJ, Reid G, Knott RB, Duff AP, Church WB. BAMLET kills chemotherapy-resistant mesothelioma cells, holding oleic acid in an activated cytotoxic state. PLoS One 2018; 13:e0203003. [PMID: 30157247 PMCID: PMC6114908 DOI: 10.1371/journal.pone.0203003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 08/13/2018] [Indexed: 12/29/2022] Open
Abstract
Malignant pleural mesothelioma is an aggressive cancer with poor prognosis. Here we have investigated in vitro efficacy of BAMLET and BLAGLET complexes (anti-cancer complexes consisting of oleic acid and bovine α-lactalbumin or β-lactoglobulin respectively) in killing mesothelioma cells, determined BAMLET and BLAGLET structures, and investigated possible biological mechanisms. We performed cell viability assays on 16 mesothelioma cell lines. BAMLET and BLAGLET having increasing oleic acid content inhibited human and rat mesothelioma cell line proliferation at decreasing doses. Most of the non-cancer primary human fibroblasts were more resistant to BAMLET than were human mesothelioma cells. BAMLET showed similar cytotoxicity to cisplatin-resistant, pemetrexed-resistant, vinorelbine-resistant, and parental rat mesothelioma cells, indicating the BAMLET anti-cancer mechanism may be different to drugs currently used to treat mesothelioma. Cisplatin, pemetrexed, gemcitabine, vinorelbine, and BAMLET, did not demonstrate a therapeutic window for mesothelioma compared with immortalised non-cancer mesothelial cells. We demonstrated by quantitative PCR that ATP synthase is downregulated in mesothelioma cells in response to regular dosing with BAMLET. We sought structural insight for BAMLET and BLAGLET activity by performing small angle X-ray scattering, circular dichroism, and scanning electron microscopy. Our results indicate the structural mechanism by which BAMLET and BLAGLET achieve increased cytotoxicity by holding increasing amounts of oleic acid in an active cytotoxic state encapsulated in increasingly unfolded protein. Our structural studies revealed similarity in the molecular structure of the protein components of these two complexes and in their encapsulation of the fatty acid, and differences in the microscopic structure and structural stability. BAMLET forms rounded aggregates and BLAGLET forms long fibre-like aggregates whose aggregation is more stable than that of BAMLET due to intermolecular disulphide bonds. The results reported here indicate that BAMLET and BLAGLET may be effective second-line treatment options for mesothelioma.
Collapse
Affiliation(s)
- Emma M. Rath
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia
| | - Yuen Yee Cheng
- Asbestos Diseases Research Institute (ADRI), Concord, NSW, Australia
- University of Sydney, Sydney, NSW, Australia
| | - Mark Pinese
- Kinghorn Cancer Centre and Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Kadir H. Sarun
- Asbestos Diseases Research Institute (ADRI), Concord, NSW, Australia
| | - Amanda L. Hudson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, University of Sydney, Sydney, NSW, Australia
| | - Christopher Weir
- Northern Blood Research Centre, Kolling Institute, University of Sydney, Sydney, NSW, Australia
| | - Yiwei D. Wang
- Burns Research, ANZAC Research Institute, Concord Hospital, University of Sydney, Concord, NSW, Australia
| | | | - Viive M. Howell
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, University of Sydney, Sydney, NSW, Australia
| | - Guo Jun Liu
- Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Rd, Lucas Heights, NSW, Australia
- Brain and Mind Centre and Faculty of Health Sciences, University of Sydney, Sydney, NSW, Australia
| | - Glen Reid
- Asbestos Diseases Research Institute (ADRI), Concord, NSW, Australia
- University of Sydney, Sydney, NSW, Australia
| | - Robert B. Knott
- Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Rd, Lucas Heights, NSW, Australia
| | - Anthony P. Duff
- Australian Nuclear Science and Technology Organisation (ANSTO), New Illawarra Rd, Lucas Heights, NSW, Australia
| | - W. Bret Church
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
33
|
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
|
34
|
Ríos-Colón L, Cajigas-Du Ross CK, Basu A, Elix C, Alicea-Polanco I, Sanchez TW, Radhakrishnan V, Chen CS, Casiano CA. Targeting the stress oncoprotein LEDGF/p75 to sensitize chemoresistant prostate cancer cells to taxanes. Oncotarget 2018; 8:24915-24931. [PMID: 28212536 PMCID: PMC5421899 DOI: 10.18632/oncotarget.15323] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 10/13/2016] [Indexed: 12/05/2022] Open
Abstract
Prostate cancer (PCa) is associated with chronic prostate inflammation resulting in activation of stress and pro-survival pathways that contribute to disease progression and chemoresistance. The stress oncoprotein lens epithelium-derived growth factor p75 (LEDGF/p75), also known as DFS70 autoantigen, promotes cellular survival against environmental stressors, including oxidative stress, radiation, and cytotoxic drugs. Furthermore, LEDGF/p75 overexpression in PCa and other cancers has been associated with features of tumor aggressiveness, including resistance to cell death and chemotherapy. We report here that the endogenous levels of LEDGF/p75 are upregulated in metastatic castration resistant prostate cancer (mCRPC) cells selected for resistance to the taxane drug docetaxel (DTX). These cells also showed resistance to the taxanes cabazitaxel (CBZ) and paclitaxel (PTX), but not to the classical inducer of apoptosis TRAIL. Silencing LEDGF/p75 effectively sensitized taxane-resistant PC3 and DU145 cells to DTX and CBZ, as evidenced by a significant decrease in their clonogenic potential. While TRAIL induced apoptotic blebbing, caspase-3 processing, and apoptotic LEDGF/p75 cleavage, which leads to its inactivation, in both taxane-resistant and -sensitive PC3 and DU145 cells, treatment with DTX and CBZ failed to robustly induce these signature apoptotic events. These observations suggested that taxanes induce both caspase-dependent and -independent cell death in mCRPC cells, and that maintaining the structural integrity of LEDGF/p75 is critical for its role in promoting taxane-resistance. Our results further establish LEDGF/p75 as a stress oncoprotein that plays an important role in taxane-resistance in mCRPC cells, possibly by antagonizing drug-induced caspase-independent cell death.
Collapse
Affiliation(s)
- Leslimar Ríos-Colón
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Christina K Cajigas-Du Ross
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Anamika Basu
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Catherine Elix
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Ivana Alicea-Polanco
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Tino W Sanchez
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Vinodh Radhakrishnan
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Chien-Shing Chen
- Department of Medicine, Division of Hematology/Medical Oncology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Carlos A Casiano
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.,Department of Medicine, Division of Rheumatology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| |
Collapse
|
35
|
Frislev HS, Boye TL, Nylandsted J, Otzen D. Liprotides kill cancer cells by disrupting the plasma membrane. Sci Rep 2017; 7:15129. [PMID: 29123177 PMCID: PMC5680231 DOI: 10.1038/s41598-017-15003-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/20/2017] [Indexed: 01/12/2023] Open
Abstract
HAMLET (human α-lactalbumin made lethal to tumour cells) is a complex of α-lactalbumin (aLA) and oleic acid (OA) which kills transformed cells, while leaving fully differentiated cells largely unaffected. Other protein-lipid complexes show similar anti-cancer potential. We call such complexes liprotides. The cellular impact of liprotides, while intensely investigated, remains unresolved. To address this, we report on the cell-killing mechanisms of liprotides prepared by incubating aLA with OA for 1 h at 20 or 80 °C (lip20 and lip80, respectively). The liprotides showed similar cytotoxicity against MCF7 cells, though lip80 acts more slowly, possibly due to intermolecular disulphide bonds formed during preparation. Liprotides are known to increase the fluidity of a membrane and transfer OA to vesicles, prompting us to focus on the effect of liprotides on the cell membrane. Extracellular Ca2+ influx is important for activation of the plasma membrane repair system, and we found that removal of Ca2+ from the medium enhanced the liprotides’ killing effect. Liprotide cytotoxicity was also increased by knockdown of Annexin A6 (ANXA6), a protein involved in plasma membrane repair. We conclude that MCF7 cells counteract liprotide-induced membrane permeabilization by activating their plasma membrane repair system, which is triggered by extracellular Ca2+ and involves ANXA6.
Collapse
Affiliation(s)
- Henriette S Frislev
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, DK-8000, Aarhus, Denmark
| | - Theresa Louise Boye
- Membrane Integrity Group, Cell Death and Metabolism Unit, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark
| | - Jesper Nylandsted
- Membrane Integrity Group, Cell Death and Metabolism Unit, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100, Copenhagen, Denmark.
| | - Daniel Otzen
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, DK-8000, Aarhus, Denmark.
| |
Collapse
|
36
|
Aqil F, Munagala R, Jeyabalan J, Agrawal AK, Gupta R. Exosomes for the Enhanced Tissue Bioavailability and Efficacy of Curcumin. AAPS JOURNAL 2017; 19:1691-1702. [PMID: 29047044 DOI: 10.1208/s12248-017-0154-9] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 09/25/2017] [Indexed: 12/31/2022]
Abstract
Exosomes are extracellular microvesicles with a particle size of 30-100 nm and carry a cargo of proteins, lipids, RNA, and DNA. Their properties of shuttling in-and-out of the cells suggest that these particles can be exploited as a nano drug carrier. In this manuscript, we show that curcumin can be delivered effectively using milk-derived exosomes. Curcumin when mixed with exosomes in the presence of 10% ethanol:acetonitrile (1:1) provided a drug load of 18-24%, and the formulation stored at - 80°C was stable for 6 months as determined by particle size analysis, drug load, and antiproliferative activity. The uptake of exosomes by cancer cells involved caveolae/clathrin-mediated endocytosis. Oral administration of exosomal curcumin (ExoCUR) in Sprague-Dawley rats demonstrated 3-5 times higher levels in various organs versus free agent. ExoCUR showed enhanced antiproliferative activity against multiple cancer cell lines including, breast, lung, and cervical cancer compared with the free curcumin. ExoCUR showed significantly higher anti-inflammatory activity measured as NF-κB activation in human lung and breast cancer cells. To determine in vivo antitumor activity, nude mice bearing the cervical CaSki tumor xenograft were treated with ExoCUR by oral gavage, curcumin diet, exosomes alone, and PBS as controls. While curcumin via dietary route failed to elicit any effect, exosomes had a modest (25-30%) tumor growth inhibition. However, ExoCUR showed significant inhibition (61%; p < 0.01) of the cervical tumor xenograft. No gross or systemic toxicity was observed in the rats administered with the exosomes or ExoCUR. These results suggest that exosomes can be developed as potential nano carriers for delivering curcumin which otherwise has encountered significant tissue bioavailability issues in the past.
Collapse
Affiliation(s)
- Farrukh Aqil
- Department of Medicine, University of Louisville, Louisville, Kentucky, 40202, USA.,James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, USA
| | - Radha Munagala
- Department of Medicine, University of Louisville, Louisville, Kentucky, 40202, USA.,James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, USA
| | - Jeyaprakash Jeyabalan
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, USA
| | - Ashish Kumar Agrawal
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, USA
| | - Ramesh Gupta
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, USA. .,Department of Pharmacology and Toxicology, University of Louisville, Delia Baxter II, Room 304E, 580 S. Preston Street, Louisville, Kentucky, 40202, USA.
| |
Collapse
|
37
|
Park Y, Kim Y, Park Y, Jin S, Hwang H, Jung YM. Formation mechanism of α-lactalabumin/oleic acid complex characterized by 2D correlation analysis. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2017; 185:93-97. [PMID: 28549296 DOI: 10.1016/j.saa.2017.05.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/14/2017] [Indexed: 06/07/2023]
Abstract
Partially unfolded α-lactalbumin (ALA) forms a complex with oleic acid (OA) that exhibits cytotoxic activity. In this study, for the first time, the pH-induced formation mechanism for ALA/OA complexes with two different molar ratios was investigated at the molecular level. For a deeper understanding of the formation mechanism of the two different ALA/OA complexes with decreasing pH, principal component analysis (PCA) and two-dimensional (2D) correlation spectroscopy were used to examine the pH-dependent IR spectra of ALA/OA complexes. By tracking the secondary structural variations in the ALA/OA complexes with decreasing pH, we successfully elucidated the formation mechanism of the ALA/OA complexes at the molecular level. The results showed that the secondary structures of theses complexes exhibited the greatest change between pH4 and pH3.5 and that the components that mainly contributed to the pH-induced transition from the N-state to the A-state were dissimilar in the two different ALA/OA complexes.
Collapse
Affiliation(s)
- Yeonju Park
- Department of Chemistry, Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yeseul Kim
- Department of Chemistry, Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yujeong Park
- Department of Chemistry, Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Sila Jin
- Department of Chemistry, Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hoon Hwang
- Department of Chemistry, Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Young Mee Jung
- Department of Chemistry, Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
38
|
Mahanta S, Prathap S, Ban DK, Paul S. Protein functionalization of ZnO nanostructure exhibits selective and enhanced toxicity to breast cancer cells through oxidative stress-based cell death mechanism. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017. [DOI: 10.1016/j.jphotobiol.2017.06.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
39
|
Sharp JA, Brennan AJ, Polekhina G, Ascher DB, Lefevre C, Nicholas KR. Dimeric but not monomeric α-lactalbumin potentiates apoptosis by up regulation of ATF3 and reduction of histone deacetylase activity in primary and immortalised cells. Cell Signal 2017; 33:86-97. [DOI: 10.1016/j.cellsig.2017.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/02/2017] [Accepted: 02/06/2017] [Indexed: 11/25/2022]
|
40
|
Munagala R, Aqil F, Jeyabalan J, Agrawal AK, Mudd AM, Kyakulaga AH, Singh IP, Vadhanam MV, Gupta RC. Exosomal formulation of anthocyanidins against multiple cancer types. Cancer Lett 2017; 393:94-102. [PMID: 28202351 PMCID: PMC5837866 DOI: 10.1016/j.canlet.2017.02.004] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/25/2017] [Accepted: 02/04/2017] [Indexed: 01/12/2023]
Abstract
Over the last two decades, berries and berry bioactives, particularly anthocyanins and their aglycones anthocyanidins (Anthos) have demonstrated excellent anti-oxidant, anti-proliferative, apoptotic and anti-inflammatory properties. However, their physicochemical and pharmacokinetic limitations such as, low permeability, and poor oral bioavailability are considered as unfavorable properties for development as drugs. Therefore there is a need to develop systems for efficient systemic delivery and robust bioavailability. In this study we prepared nano-formulation of bilberry-derived Anthos using exosomes harvested from raw bovine milk. Exosomal formulation of Anthos enhanced antiproliferative and anti-inflammatory effects compared with the free Anthos against various cancer cells in vitro. Our data also showed significantly enhanced therapeutic response of exosomal-Anthos formulation compared with the free Anthos against lung cancer tumor xenograft in nude mice. The Anthos showed no signs of gross or systemic toxicity in wild-type mice. Thus, exosomes provide an effective alternative for oral delivery of Anthos that is efficacious, cost-effective, and safe, and this regimen can be developed as a non-toxic, widely applicable therapeutic agent.
Collapse
Affiliation(s)
- Radha Munagala
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA; Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Farrukh Aqil
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA; Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Jeyaprakash Jeyabalan
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Ashish K Agrawal
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Ashley M Mudd
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Al Hassan Kyakulaga
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - Inder P Singh
- National Institute of Pharmaceutical Education and Research, S.A.S Nagar, India
| | - Manicka V Vadhanam
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Ramesh C Gupta
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
41
|
Agrawal AK, Aqil F, Jeyabalan J, Spencer WA, Beck J, Gachuki BW, Alhakeem SS, Oben K, Munagala R, Bondada S, Gupta RC. Milk-derived exosomes for oral delivery of paclitaxel. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:1627-1636. [PMID: 28300659 DOI: 10.1016/j.nano.2017.03.001] [Citation(s) in RCA: 379] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/13/2017] [Accepted: 03/03/2017] [Indexed: 12/11/2022]
Abstract
In this report milk-derived exosomes have been investigated for oral delivery of the chemotherapeutic drug paclitaxel (PAC) as an alternative to conventional i.v. therapy for improved efficacy and reduced toxicity. PAC-loaded exosomes (ExoPAC) were found to have a particle size of ~108 nm, a narrow particle size distribution (PDI ~0.190), zeta potential (~ -7 mV) and a practical loading efficiency of ~8%. Exosomes and ExoPAC exhibited excellent stability in the presence of simulated-gastrointestinal fluids, and during the storage at -80 °C. A sustained release of PAC was also observed up to 48 h in vitro using PBS (pH 6.8). Importantly, ExoPAC delivered orally showed significant tumor growth inhibition (60%; P<0.001) against human lung tumor xenografts in nude mice. Treatment with i.p. PAC at the same dose as ExoPAC, however, showed modest but statistically insignificant inhibition (31%). Moreover, ExoPAC demonstrated remarkably lower systemic and immunologic toxicities as compared to i.v. PAC.
Collapse
Affiliation(s)
- Ashish K Agrawal
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY
| | - Farrukh Aqil
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY; Department of Medicine, University of Louisville, Louisville, KY
| | | | | | | | - Beth W Gachuki
- Department of Microbiology, Immunology & Molecular Genetics, and Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Sara S Alhakeem
- Department of Microbiology, Immunology & Molecular Genetics, and Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Karine Oben
- Department of Microbiology, Immunology & Molecular Genetics, and Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Radha Munagala
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY; Department of Medicine, University of Louisville, Louisville, KY
| | - Subbarao Bondada
- Department of Microbiology, Immunology & Molecular Genetics, and Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Ramesh C Gupta
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY; 3P Biotechnologies, Inc., Louisville, KY; Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY.
| |
Collapse
|
42
|
Fang B, Zhang M, Wu H, Fan X, Ren F. Internalization properties of the anti-tumor α-lactalbumin-oleic acid complex. Int J Biol Macromol 2017; 96:44-51. [DOI: 10.1016/j.ijbiomac.2016.12.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 11/30/2016] [Accepted: 12/12/2016] [Indexed: 10/20/2022]
|
43
|
INAGAKI M, KAWAI S, IJIER X, FUKUOKA M, YABE T, IWAMOTO S, KANAMARU Y. Effects of heat treatment on conformation and cell growth activity of alpha- lactalbumin and beta-lactoglobulin from market milk . Biomed Res 2017; 38:53-59. [DOI: 10.2220/biomedres.38.53] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Mizuho INAGAKI
- Department of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University
| | - Shuji KAWAI
- Department of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University
- Deceased
| | - X IJIER
- Department of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University
| | - Mayuko FUKUOKA
- Department of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University
| | - Tomio YABE
- Department of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University
| | - Satoshi IWAMOTO
- Department of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University
| | - Yoshihiro KANAMARU
- Department of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University
| |
Collapse
|
44
|
A Characeae Cells Plasma Membrane as a Model for Selection of Bioactive Compounds and Drugs: Interaction of HAMLET-Like Complexes with Ion Channels of Chara corallina Cells Plasmalemma. J Membr Biol 2016; 249:801-811. [DOI: 10.1007/s00232-016-9930-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 09/07/2016] [Indexed: 01/10/2023]
|
45
|
Seebacher N, Lane DJR, Richardson DR, Jansson PJ. Turning the gun on cancer: Utilizing lysosomal P-glycoprotein as a new strategy to overcome multi-drug resistance. Free Radic Biol Med 2016; 96:432-45. [PMID: 27154979 DOI: 10.1016/j.freeradbiomed.2016.04.201] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 04/01/2016] [Accepted: 04/29/2016] [Indexed: 01/02/2023]
Abstract
Oxidative stress plays a role in the development of drug resistance in cancer cells. Cancer cells must constantly and rapidly adapt to changes in the tumor microenvironment, due to alterations in the availability of nutrients, such as glucose, oxygen and key transition metals (e.g., iron and copper). This nutrient flux is typically a consequence of rapid growth, poor vascularization and necrosis. It has been demonstrated that stress factors, such as hypoxia and glucose deprivation up-regulate master transcription factors, namely hypoxia inducible factor-1α (HIF-1α), which transcriptionally regulate the multi-drug resistance (MDR), transmembrane drug efflux transporter, P-glycoprotein (Pgp). Interestingly, in addition to the established role of plasma membrane Pgp in MDR, a new paradigm of intracellular resistance has emerged that is premised on the ability of lysosomal Pgp to transport cytotoxic agents into this organelle. This mechanism is enabled by the topological inversion of Pgp via endocytosis resulting in the transporter actively pumping agents into the lysosome. In this way, classical Pgp substrates, such as doxorubicin (DOX), can be actively transported into this organelle. Within the lysosome, DOX becomes protonated upon acidification of the lysosomal lumen, causing its accumulation. This mechanism efficiently traps DOX, preventing its cytotoxic interaction with nuclear DNA. This review discusses these effects and highlights a novel mechanism by which redox-active and protonatable Pgp substrates can utilize lysosomal Pgp to gain access to this compartment, resulting in catastrophic lysosomal membrane permeabilization and cell death. Hence, a key MDR mechanism that utilizes Pgp (the "gun") to sequester protonatable drug substrates safely within lysosomes can be "turned on" MDR cancer cells to destroy them from within.
Collapse
Affiliation(s)
- Nicole Seebacher
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Blackburn Building (D06), University of Sydney, Sydney, New South Wales 2006, Australia
| | - Darius J R Lane
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Blackburn Building (D06), University of Sydney, Sydney, New South Wales 2006, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Blackburn Building (D06), University of Sydney, Sydney, New South Wales 2006, Australia
| | - Patric J Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Blackburn Building (D06), University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
46
|
Aits S, Kricker J, Liu B, Ellegaard AM, Hämälistö S, Tvingsholm S, Corcelle-Termeau E, Høgh S, Farkas T, Holm Jonassen A, Gromova I, Mortensen M, Jäättelä M. Sensitive detection of lysosomal membrane permeabilization by lysosomal galectin puncta assay. Autophagy 2016; 11:1408-24. [PMID: 26114578 DOI: 10.1080/15548627.2015.1063871] [Citation(s) in RCA: 306] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lysosomal membrane permeabilization (LMP) contributes to tissue involution, degenerative diseases, and cancer therapy. Its investigation has, however, been hindered by the lack of sensitive methods. Here, we characterize and validate the detection of galectin puncta at leaky lysosomes as a highly sensitive and easily manageable assay for LMP. LGALS1/galectin-1 and LGALS3/galectin-3 are best suited for this purpose due to their widespread expression, rapid translocation to leaky lysosomes and availability of high-affinity antibodies. Galectin staining marks individual leaky lysosomes early during lysosomal cell death and is useful when defining whether LMP is a primary or secondary cause of cell death. This sensitive method also reveals that cells can survive limited LMP and confirms a rapid formation of autophagic structures at the site of galectin puncta. Importantly, galectin staining detects individual leaky lysosomes also in paraffin-embedded tissues allowing us to demonstrate LMP in tumor xenografts in mice treated with cationic amphiphilic drugs and to identify a subpopulation of lysosomes that initiates LMP in involuting mouse mammary gland. The use of ectopic fluorescent galectins renders the galectin puncta assay suitable for automated screening and visualization of LMP in live cells and animals. Thus, the lysosomal galectin puncta assay opens up new possibilities to study LMP in cell death and its role in other cellular processes such as autophagy, senescence, aging, and inflammation.
Collapse
Affiliation(s)
- Sonja Aits
- a Cell Death and Metabolism Unit; Center for Autophagy, Recycling and Disease; Danish Cancer Society Research Center ; Copenhagen , Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Basu A, Cajigas-Du Ross CK, Rios-Colon L, Mediavilla-Varela M, Daniels-Wells TR, Leoh LS, Rojas H, Banerjee H, Martinez SR, Acevedo-Martinez S, Casiano CA. LEDGF/p75 Overexpression Attenuates Oxidative Stress-Induced Necrosis and Upregulates the Oxidoreductase ERP57/PDIA3/GRP58 in Prostate Cancer. PLoS One 2016; 11:e0146549. [PMID: 26771192 PMCID: PMC4714844 DOI: 10.1371/journal.pone.0146549] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 12/19/2015] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer (PCa) mortality is driven by highly aggressive tumors characterized by metastasis and resistance to therapy, and this aggressiveness is mediated by numerous factors, including activation of stress survival pathways in the pro-inflammatory tumor microenvironment. LEDGF/p75, also known as the DFS70 autoantigen, is a stress transcription co-activator implicated in cancer, HIV-AIDS, and autoimmunity. This protein is targeted by autoantibodies in certain subsets of patients with PCa and inflammatory conditions, as well as in some apparently healthy individuals. LEDGF/p75 is overexpressed in PCa and other cancers, and promotes resistance to chemotherapy-induced cell death via the transactivation of survival proteins. We report in this study that overexpression of LEDGF/p75 in PCa cells attenuates oxidative stress-induced necrosis but not staurosporine-induced apoptosis. This finding was consistent with the observation that while LEDGF/p75 was robustly cleaved in apoptotic cells into a p65 fragment that lacks stress survival activity, it remained relatively intact in necrotic cells. Overexpression of LEDGF/p75 in PCa cells led to the upregulation of transcript and protein levels of the thiol-oxidoreductase ERp57 (also known as GRP58 and PDIA3), whereas its depletion led to ERp57 transcript downregulation. Chromatin immunoprecipitation and transcription reporter assays showed LEDGF/p75 binding to and transactivating the ERp57 promoter, respectively. Immunohistochemical analysis revealed significantly elevated co-expression of these two proteins in clinical prostate tumor tissues. Our results suggest that LEDGF/p75 is not an inhibitor of apoptosis but rather an antagonist of oxidative stress-induced necrosis, and that its overexpression in PCa leads to ERp57 upregulation. These findings are of significance in clarifying the role of the LEDGF/p75 stress survival pathway in PCa.
Collapse
Affiliation(s)
- Anamika Basu
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350, United States of America
- * E-mail:
| | - Christina K. Cajigas-Du Ross
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350, United States of America
| | - Leslimar Rios-Colon
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350, United States of America
| | - Melanie Mediavilla-Varela
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350, United States of America
| | - Tracy R. Daniels-Wells
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350, United States of America
| | - Lai Sum Leoh
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350, United States of America
| | - Heather Rojas
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, California 92350, United States of America
| | - Hiya Banerjee
- Novartis Pharmaceutical Oncology, East Hanover, New Jersey 08807, United States of America
| | - Shannalee R. Martinez
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350, United States of America
| | - Stephanny Acevedo-Martinez
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350, United States of America
| | - Carlos A. Casiano
- Center for Health Disparities and Molecular Medicine, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California 92350, United States of America
- Department of Medicine, Loma Linda University School of Medicine, Loma Linda, California 92350, United States of America
| |
Collapse
|
48
|
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: 1.7] [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
|
49
|
Groth-Pedersen L, Jäättelä M, Nylandsted J. A Method to Monitor Lysosomal Membrane Permeabilization by Immunocytochemistry. Cold Spring Harb Protoc 2015; 2015:904-907. [PMID: 26430254 DOI: 10.1101/pdb.prot086181] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Programmed cell death involving lysosomal membrane permeabilization (LMP) is a common phenomenon--more the rule than the exception under various cytotoxic stimuli and stressful cellular conditions. The protocol presented here is based on immunocytochemical staining of cathepsin B or L to visualize translocation from the lysosomal lumen to the cytosol. In healthy cells, cathepsins appear in localized punctate structures representing intact lysosomes, whereas LMP results in a diffuse staining pattern throughout the cytoplasm. LMP can be triggered upstream, downstream, or independently of the classical apoptotic death pathway involving mitochondrial outer membrane permeabilization (MOMP). Co-staining with antibodies recognizing the active form of Bax allows investigation of the order of events between LMP and MOMP in death signaling.
Collapse
Affiliation(s)
- Line Groth-Pedersen
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet University Hospital, DK-2100 Copenhagen, Denmark; Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
| | - Marja Jäättelä
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
| | - Jesper Nylandsted
- Unit for Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
| |
Collapse
|
50
|
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: 0.9] [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
|