Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 290
Filter
1.
Int J Pharm ; 663: 124578, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39153643

ABSTRACT

Interruption of wound healing by multi-drug resistant-bacterial infection is a harmful issue for the worldwide health care system, and conventional treatment approaches may not resolve this issue due to antimicrobial resistance. So, there is an unmet need to develop scaffolds with intrinsic wound healing properties to combat bacterial-infected wounds. Inspired by the α-lactalbumin's (Lalb's) ability to promote collagen synthesis, we herein electrospun Lalb with cephalexin (CPL) and epigallocatechin (EP) to produce nanofibers (CE-Lalb NFs) to solve this issue. The CE-Lalb NFs were prepared using the electrospinning technique and subjected to physicochemical characterizations, in vitro, and in vivo assessments. The CE-Lalb NFs promoted fibroblast migration, proliferation, and collagen synthesis, while CPL/EP annihilated MRSA and E. coli infections. Physicochemical characterizations proved the successful fabrication and doping of CE-Lalb NFs. Antimicrobial assays and fractional inhibitory concentration index (FICI) declared synergistic antibacterial activity of CE-Lalb NFs against MRSA and E. coli. The in vivo and immunohistochemical data evidenced its exceptional potential for wound healing, promoting growth factor, collagen synthesis, and reduced scar formation. The presence of mature collagen, fewer inflammatory cytokines, increased expression of blood vessels, and low expression of IL-6 at the wound site support in vitro and in vivo results. In our view, the tailored scaffold is the next step for personalized wound dressings that could meet patients with infected wounds' unmet needs by the subscription of noninvasive and easily navigable therapeutic options.


Subject(s)
Anti-Bacterial Agents , Lactalbumin , Wound Healing , Wound Healing/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/administration & dosage , Lactalbumin/chemistry , Lactalbumin/pharmacology , Tissue Scaffolds/chemistry , Escherichia coli/drug effects , Mice , Nanofibers/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Collagen , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/pharmacology , Catechin/administration & dosage , Male , Cephalexin/pharmacology , Cephalexin/chemistry , Cephalexin/administration & dosage , Fibroblasts/drug effects , Regeneration/drug effects , Humans , Cell Proliferation/drug effects , Escherichia coli Infections/drug therapy , Staphylococcal Infections/drug therapy , Cell Movement/drug effects , Rats
2.
Mol Nutr Food Res ; 67(16): e2200499, 2023 08.
Article in English | MEDLINE | ID: mdl-37354055

ABSTRACT

SCOPE: Dietary intervention has emerged as a promising strategy for the management of nonalcoholic fatty liver disease (NAFLD). The aim of this study is to investigate the ameliorative effects of the α-lactalbumin peptide Asp-Gln-Trp (DQW) against NAFLD and the underlying mechanism. METHODS AND RESULTS: The models of lipid metabolism disorders are established both in HepG2 cells and in C57BL/6J mice. The results demonstrate that DQW activates peroxisome proliferator-activated receptor α (PPARα) and subsequently ameliorates lipid deposition and oxidative stress in vitro. Interestingly, GW6471 markedly attenuates the modulatory effects of DQW on the PPARα pathway in HepG2 cells. Moreover, results of in vivo experiments indicate that DQW alleviates body weight gain, dyslipidemia, hepatic steatosis, and oxidative stress in high-fat-diet (HFD)-induced NAFLD mice. At the molecular level, DQW activates PPARα, subsequently enhances fatty acid ß-oxidation, and reduces lipogenesis, thereby ameliorating hepatic steatosis. Meanwhile, DQW may ameliorate liver injury and oxidative stress via activating the PPARα/nuclear-factor erythroid 2 (Nrf2)/heme-oxygenase 1 (HO-1) pathway. CONCLUSION: Those results indicate that α-lactalbumin peptide DQW may be an effective dietary supplement for alleviating NAFLD by alleviating lipid deposition and oxidative stress.


Subject(s)
Non-alcoholic Fatty Liver Disease , Mice , Animals , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , PPAR alpha/metabolism , Fatty Acids, Nonesterified/pharmacology , Lactalbumin/pharmacology , Lactalbumin/metabolism , Hep G2 Cells , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Liver/metabolism , Oxidative Stress , Lipid Metabolism
3.
J Dairy Sci ; 106(4): 2271-2288, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36797178

ABSTRACT

The objective of this study was to investigate the mechanism by which the α-lactalbumin peptides Gly-Ile-Asn-Tyr (GINY) and Asp-Gln-Trp (DQW) ameliorate free fatty acid-induced lipid deposition in HepG2 cells. The results show that GINY and DQW reduced triglyceride, total cholesterol, and free fatty acid levels significantly in free fatty acid-treated HepG2 cells. Based on proteomic analysis, GINY and DQW alleviated lipid deposition and oxidative stress mainly through the peroxisome proliferator-activated receptor (PPAR) pathway, fatty acid metabolism, oxidative phosphorylation, and response to oxidative stress. In vitro experiments confirmed that GINY and DQW upregulated the mRNA and protein expression of fatty acid ß-oxidation-related and oxidative stress-related genes, and downregulated the mRNA and protein expression of lipogenesis-related genes by activating peroxisome proliferator-activated receptor α (PPARα). Meanwhile, GINY and DQW reduced free fatty acid-induced lipid droplet accumulation and reactive oxygen species generation, and enhanced the mitochondrial membrane potential and ATP levels. Furthermore, GINY and DQW enhanced carnitine palmitoyl-transferase 1a (CPT-1a) and superoxide dismutase activities, and diminished acetyl-coenzyme A carboxylase 1 (ACC1) and fatty acid synthase (FASN) activities in a PPARα-dependent manner. Interestingly, GW6471 (a PPARα inhibitor) weakened the effects of GINY and DQW on the PPARα pathway. Hence, our findings suggest that GINY and DQW have the potential to alleviate nonalcoholic fatty liver disease by activating the PPARα pathway.


Subject(s)
Lactalbumin , Non-alcoholic Fatty Liver Disease , Animals , Humans , Hep G2 Cells , Lactalbumin/pharmacology , Lactalbumin/metabolism , PPAR alpha/genetics , Fatty Acids, Nonesterified/metabolism , Proteomics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/veterinary , Oxidative Stress , Lipid Metabolism , Peptides/pharmacology , Peptides/metabolism , RNA, Messenger/metabolism , Liver/metabolism
4.
Appetite ; 180: 106339, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36216216

ABSTRACT

Protein is considered to be the most satiating food macronutrient and the satiating effect may be dependent on the source of the protein. The maize-derived protein zein and milk protein casein have been shown previously to lower stomach emptying rate more than dairy whey protein, but the effect of zein on satiety has not been evaluated. The objective was to compare the satiating effects of zein and casein, with whey protein and its protein component α-lactalbumin. The study was a randomised crossover design with thirteen normal-weight men (mean age 27.8 years and mean BMI 24.4 kg/m2) consuming isoenergetic (∼4000 kJ, ∼990 kcal) preload mixed meals enriched with Zein, Casein, whey protein isolate (Whey), α-lactalbumin (ALac), or maltodextrin carbohydrate (Carb). Consumption of an ad libitum standardised test meal of chicken fried rice and water provided 360 min following ingestion of the preload meal was measured, and subjective feelings of appetite (hunger, fullness, desire to eat, and prospective food consumption) were assessed using 100-mm visual analogue scales (VAS). There were no differences among the five preload mixed meals in the amount of chicken fried rice consumed at the ad libitum test meal (mean ± sem: 531.6 ± 35.0 g, p = 0.47) or total (preload + test meal) energy intakes (mean ± sem: 5780.5 ± 146.0 kJ, p = 0.29). The subjective VAS appetite ratings and total area under the curve responses for hunger, fullness, desire to eat, and prospective food consumption, were not different following consumption of all five preload mixed meals (p > 0.05). The findings indicate that the effects of zein and casein on satiety were not different from the satiating effects of whey protein and α-lactalbumin.


Subject(s)
Eating , Milk Proteins , Satiation , Whey Proteins , Zein , Caseins/pharmacology , Lactalbumin/pharmacology , Whey Proteins/pharmacology , Zea mays , Cross-Over Studies , Humans , Male , Adult
5.
Eur J Appl Physiol ; 123(2): 381-393, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36443490

ABSTRACT

PURPOSE: The neurotransmitter serotonin has a strong effect on behaviour and motor control. Regarding motor control, serotonin contributes to the development of fatigue and is also involved in the ability of motor neurones to operate across a large range of forces (gain control). The consumption of tryptophan-rich supplements (such as α-lactalbumin) is of interest because this amino acid is the only precursor for brain serotonin synthesis. Therefore, the purpose of this study was to determine the effects of α-lactalbumin supplementation on neuromuscular performance. METHODS: Using a randomised double-blind cross-over design, 16 healthy participants performed plantar flexor and handgrip maximal voluntary contractions, a 30-s submaximal handgrip contraction, and a plantar flexor fatigue protocol before and 90 min after consuming either 40 g of α-lactalbumin, an isonitrogenous beverage (Zein) or an isocaloric beverage (corn-starch). Sleepiness, mood, and cognition were assessed to evaluate any psychological effects. RESULTS: α-Lactalbumin decreased force steadiness by 25% during the sustained submaximal handgrip contraction (p < 0.01) and induced greater fatigue (15% reduction in total torque-time integral, p = 0.01) during the fatigue protocol. These effects were not observed for the other control beverages. No effects were found for maximal or explosive strength, or psychological measurements. CONCLUSIONS: 40 g of α-lactalbumin increased handgrip force variability and reduced performance during fatiguing muscle contractions but did not influence brief maximal contractions or psychological parameters in healthy individuals. These findings support the hypothesis that the consumption of α-lactalbumin can increase motor neurone input-output gain and exacerbate central fatigue during sustained maximal exercise.


Subject(s)
Lactalbumin , Muscle Fatigue , Humans , Lactalbumin/pharmacology , Cross-Over Studies , Muscle Fatigue/physiology , Hand Strength , Serotonin , Muscle Contraction , Fatigue , Electromyography/methods , Muscle, Skeletal/physiology , Isometric Contraction/physiology
6.
Eur J Appl Physiol ; 123(2): 395-404, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36443491

ABSTRACT

INTRODUCTION: We tested two strategies that hypothetically increase serotonin availability (α-lactalbumin consumption and a remote submaximal handgrip contraction) on estimates of persistent inward currents (PICs) amplitude of soleus muscle in healthy participants. METHODS: With a randomised, double-blind, and cross-over design, 13 healthy participants performed triangular-shaped ramp contractions with their plantar flexors (20% of maximal torque), followed by a 30-s handgrip sustained contraction (40% of maximal force) and consecutive repeated triangular-shaped contractions. This was performed before and after the consumption of either 40 g of α-lactalbumin, an isonitrogenous beverage (Zein) or an isocaloric beverage (Corn-starch). Soleus motor units discharge rates were analysed from high-density surface electromyography signals. PICs were estimated by calculating the delta frequency (ΔF) of motor unit train spikes using the paired motor unit technique. RESULTS: ΔF (0.19 pps; p = 0.001; d = 0.30) and peak discharge rate (0.20 pps; p < 0.001; d = 0.37) increased after the handgrip contraction, irrespective of the consumed supplement. No effects of α-lactalbumin were observed. CONCLUSIONS: Our results indicate that 40 g of α-lactalbumin was unable to modify intrinsic motoneuron excitability. However, performing a submaximal handgrip contraction before the plantar flexion triangular contraction was capable of increasing ΔF and discharge rates on soleus motor units. These findings highlight the diffused effects of serotonergic input, its effects on motoneuron discharge behaviour, and suggest a cross-effector effect within human motoneurons.


Subject(s)
Hand Strength , Lactalbumin , Humans , Lactalbumin/pharmacology , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Electromyography/methods , Motor Neurons/physiology , Isometric Contraction/physiology
7.
Int Immunopharmacol ; 113(Pt A): 109291, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36252495

ABSTRACT

Cardiovascular aging is the most important factor leading to cardiovascular disease (CVD), and the incidence and severity of cardiovascular events increase with age. Cardiovascular disease is one of the leading causes of death in the aging population. Therefore, it is extremely urgent to develop and explore effective drugs or bioactive molecules to prevent cardiovascular aging and related diseases. In the current work, the effect of bovine α-lactalbumin (α-lactalbumin is one of the major bioactive protein molecules in milk) on cardiovascular aging was investigated in vitro and in vivo. First, a cellular model of cardiovascular aging was established using H2O2-induced in vitro cellular models. It was found that α-lactalbumin could alleviate cardiovascular senescence by assessing Sa-ß-gal and senescence-related markers (such as p16/p21/p53) in in vitro cellular models. Bovine α-lactalbumin attenuated aging-related inflammation and oxidative stress. Furthermore, aged mice were used as an in vivo cardiovascular aging model. We explored the effect of α-lactalbumin on cardiovascular aging and found that cardiovascular aging was significantly attenuated by evaluating Sa-ß-gal staining and aging-related marker molecules. Mechanistically, we found that α-lactalbumin may alleviate cardiovascular aging by regulating the expression of Sirt1 (Sirtuin 1). In summary, in the current work, we systematically explored the potential biological activity of α-lactalbumin against cardiovascular aging and found that α-lactalbumin has good anti-aging potential in vitro and in vivo, suggesting that α-lactalbumin could be used as an antiaging functional food in the future.


Subject(s)
Cardiovascular Diseases , Lactalbumin , Mice , Cattle , Animals , Lactalbumin/pharmacology , Lactalbumin/therapeutic use , Cellular Senescence , Hydrogen Peroxide/pharmacology , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/prevention & control , Aging , Sirtuin 1/metabolism
8.
Food Chem ; 397: 133820, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-35932687

ABSTRACT

Food contamination and poisoning caused by bacteria will endanger human health, and the development of natural antibacterial agents is a pressing issue. We prepared ALA-Car complex and demonstrated its formation by multi-spectroscopy techniques and localized surface plasmon resonance experiments. Computer simulations have shown that van der Waals forces dominate the interaction between ALA and Car. The minimum inhibitory concentration (MIC) of Car toward Gram-negative Escherichia coli was decreased from 336 µg/mL to 224 µg/mL after binding to ALA. It had little effect on the MIC of Gram-positive Staphylococcus aureus (224 µg/mL), but further proved Car had a weaker antibacterial activity than the ALA-Car complex by the spread plate method. Overall, this work demonstrated that the ALA-Car complex had significantly higher antibacterial activities than Car, further advancing the development of natural antibacterial agents.


Subject(s)
Anti-Bacterial Agents , Lactalbumin , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Cymenes , Humans , Lactalbumin/pharmacology , Microbial Sensitivity Tests
9.
Biofactors ; 48(5): 1145-1159, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35388547

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.


Subject(s)
Lipid Bilayers , Urinary Bladder Neoplasms , Humans , Lactalbumin/chemistry , Lactalbumin/metabolism , Lactalbumin/pharmacology , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Oleic Acid/chemistry , Oleic Acid/metabolism , Oleic Acid/pharmacology , Phosphatidylcholines/chemistry , Sphingomyelins/chemistry , Tissue Extracts , Unilamellar Liposomes
10.
ACS Chem Neurosci ; 13(8): 1263-1272, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35385250

ABSTRACT

Parkinson's disease (PD) is the second most common devastating neurodegenerative disorder. Presently used therapies for PD have severe side effects and are limited to only temporary improvement. Therefore, a new therapeutic approach to treat PD urgently needs to be developed. α-Lactalbumin, the most abundant milk protein in camel milk, has been attributed to various medicinal properties. This study intended to investigate the neuroprotective efficacy of the camel α-lactalbumin and oleic acid (CLOA) complex. One mechanism postulated to underlie neuroprotection by the CLOA complex is the induction of silent information regulatory protein (SIRT1). SIRT1 is known to be involved in several pathological and physiological processes, and it has been suggested that SIRT1 plays a protective role in PD. Oxidative stress, inflammation, mitochondrial dysfunction, and apoptosis are involved in PD pathogenesis. Our results revealed that SIRT1 inhibits oxidative stress by maintaining HIF-1α in a deacetylated state. SIRT1 upregulates the expression of FOXO3a and HSF-1, thus inhibiting apoptosis and maintaining the homeostasis of cellular proteins. Increased SIRT1 expression reduces the levels of TNF-α, IL-6, and IL-8, which in turn inhibits neuroinflammation. In addition to SIRT1, the CLOA complex also enhances the expression of survivin and leptin and promotes the survival of neuroblastoma cells. Altogether, our results suggest that the CLOA complex might be a novel therapeutic molecule that could ameliorate neuronal cell damage in PD.


Subject(s)
Neuroprotective Agents , Parkinson Disease , Animals , Camelus/metabolism , Lactalbumin/metabolism , Lactalbumin/pharmacology , Lactalbumin/therapeutic use , Neuroprotection , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oleic Acid/pharmacology , Oleic Acid/therapeutic use , Oxidative Stress , Parkinson Disease/drug therapy , Rotenone , Sirtuin 1/metabolism , Sirtuin 1/pharmacology , Sirtuin 1/therapeutic use
11.
Food Funct ; 13(4): 2323-2335, 2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35142310

ABSTRACT

Bioactive peptides in bovine α-lactalbumin were isolated and identified, and the effects and mechanisms of peptide KILDK on insulin resistance in 3T3-L1 adipocytes were investigated. Mature 3T3-L1 adipocytes were stimulated with TNF-α to induce insulin resistance. Bovine α-lactalbumin hydrolysates (α-LAH) were subjected to stimulated gastrointestinal digestion and Caco-2 absorption, and GD-α-LAH and CA-α-LAH were obtained. Our results demonstrated that α-LAH, GD-α-LAH, and CA-α-LAH increased glucose uptake, enhanced Akt phosphorylation (Ser473), and decreased IRS-1 phosphorylation (Ser307) in insulin resistant 3T3-L1 adipocytes. Gel filtration chromatography and liquid chromatography-electrospray ionization tandem mass spectrometry (LC-ESI MS/MS) were used to separate and identify bioactive peptides. The identified peptide KILDK attenuated insulin resistance in 3T3-L1 adipocytes, which was attributed to the suppression of JNK phosphorylation (Thr183/Tyr185). Moreover, KILDK downregulated pro-inflammatory genes through blocking NF-κB signaling. Our findings suggested that bovine α-LAH might be a potential ingredient against insulin resistance.


Subject(s)
Inflammation/prevention & control , Insulin Resistance , Lactalbumin/pharmacology , Peptides/pharmacology , 3T3-L1 Cells/drug effects , Animals , Caco-2 Cells/drug effects , Cattle , Humans , Inflammation/chemically induced , MAP Kinase Kinase 4/metabolism , Mice , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha
12.
Neuropharmacology ; 198: 108782, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34492286

ABSTRACT

The Microbiota-Gut-Brain axis (MGBA) is a bidirectional communication pathway between gut bacteria and the central nervous system (CNS) (including the intestine) that exerts a profound influence on neural development, neuroinflammation, activation of stress response and neurotransmission, in addition to modulating complex behaviours, such as sociability and anxiety. Several MGBA modulating approaches are possible, such as probiotic administration. A reasonable pharmacological approach would also be the contemporarily administration of both prebiotics and postbiotics. To test this hypothesis, we probed the effects of α-lactalbumin (ALAC; a prebiotic in the dose range of 125-500 mg/kg) and sodium butyrate (NaB; a postbiotic in the dose range of 30-300 mg/kg) alone and in combination. We used two animal behavioural models of idiopathic autism, (BTBR mice) and anxiety/depression (chronic unexpected mild stress - CUMS mice) respectively, using several standard behavioural paradigms such as Three-chamber social interaction test, Marble burying assay, depression-, anxiety- and memory-tests. In BTBR autistic mice, we found that both ALAC and NaB improve animal sociability, and memory in the passive avoidance (PA); drug combination was more effective in almost all tests also reducing immobility time in the forced swimming test (FST), which was not affected by single drug administration. Similarly, in the CUMS mice, single drug administration was effective in improving: 1) depressive-like behaviour in the FST and sucrose preference test; 2) memory and learning in the PA, novel object recognition and Morris water maze tests. Drug combination was again more effective than single drug administration in most cases; however, in the CUMS model, neither single drug or combination was effective in the elevated plus maze test for anxiety. Our results suggest that in both models, ALAC and NaB combination is more effective in improving some pathological aspects of animal behaviour than single administration and that the prebiotic/postbiotic approach should be considered a reasonable approach for the manipulation of the MGBA to improve efficacy.


Subject(s)
Autistic Disorder/prevention & control , Brain-Gut Axis , Depression/prevention & control , Gastrointestinal Microbiome , Prebiotics , Animals , Anxiety/psychology , Autistic Disorder/psychology , Avoidance Learning/drug effects , Behavior, Animal , Butyric Acid/pharmacology , Depression/psychology , Disease Models, Animal , Drug Synergism , Humans , Lactalbumin/pharmacology , Male , Memory , Mice , Mice, Inbred C57BL , Social Behavior , Stress, Psychological/psychology , Swimming/psychology
13.
Int J Biol Macromol ; 187: 513-527, 2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34310992

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.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Lactalbumin/pharmacology , Leukemia/drug therapy , NADPH Oxidase 4/metabolism , Protein Phosphatase 2/metabolism , Semicarbazides/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Calcium Signaling/drug effects , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Female , Humans , Leukemia/enzymology , Leukemia/genetics , Leukemia/pathology , MCF-7 Cells , NADPH Oxidase 4/genetics , Protein Phosphatase 2/genetics , Proteolysis , Reactive Oxygen Species/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Tristetraprolin/metabolism , Tumor Necrosis Factor-alpha/genetics , U937 Cells , p38 Mitogen-Activated Protein Kinases/metabolism
14.
J Dairy Res ; 88(2): 221-225, 2021 May.
Article in English | MEDLINE | ID: mdl-33985600

ABSTRACT

Alpha-lactalbumin (α-LA) and ß-lactoglobulin (ß-LG) are contained in bovine milk whey. Chemical and physical treatments are known to alter the conformation of these proteins and we have previously reported that α-LA denatured with trifluoroethanol (TFE) and isolated from sterilized market milk inhibited the growth of rat crypt IEC-6 cells. In the present study, we aimed to evaluate the effects of TFE-treated α-LA and ß-LG on cell growth using cultured intestinal cells and on their safety using a suckling mouse model. First, we investigated the effect of the TFE-treated whey proteins on human colonic Caco-2 cells at various differentiation stages. In the undifferentiated stage, we assessed cell growth by a water-soluble tetrazolium-1 method. The native whey proteins enhanced cell proliferation, whereas the TFE-treated whey proteins strongly inhibited cell growth. We investigated cell barrier function in the post-differentiated stage by measuring transepithelial electrical resistance (TER). Not only native but also the TFE-treated whey proteins increased TER. Next, we evaluated whether the TFE-treated α-LA and ß-LG have adverse effects on healthy suckling mice. No mice given by the TFE-treated samples showed any adverse symptoms. We also performed a safety test using a human rotavirus infected gastrointestinal disease suckling mice model. Even the TFE-treated whey proteins appeared to prevent the development of diarrheal symptoms without any adverse effects. Although we cannot know the effect of long-term ingestion of denatured whey proteins, these results suggest that they have no adverse effects on differentiated intestinal cells and digestive tract, at least in short-term ingestion.


Subject(s)
Gastrointestinal Tract/drug effects , Protein Denaturation , Whey Proteins/chemistry , Whey Proteins/pharmacology , Animals , Animals, Suckling , Caco-2 Cells , Cattle , Cell Differentiation , Cell Line , Cell Proliferation/drug effects , Diarrhea/drug therapy , Humans , Lactalbumin/chemistry , Lactalbumin/pharmacology , Lactoglobulins/chemistry , Lactoglobulins/pharmacology , Mice , Mice, Inbred BALB C , Trifluoroethanol/chemistry
15.
Eur J Cancer Prev ; 30(2): 178-187, 2021 03 01.
Article in English | MEDLINE | ID: mdl-32694279

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.


Subject(s)
Antineoplastic Agents , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma, Squamous Cell/drug therapy , Cattle , Humans , Lactalbumin/pharmacology , Mouth Neoplasms/drug therapy , Oleic Acid/pharmacology , Squamous Cell Carcinoma of Head and Neck , Tumor Suppressor Protein p53/genetics
16.
Eur J Immunol ; 51(4): 965-977, 2021 04.
Article in English | MEDLINE | ID: mdl-33348422

ABSTRACT

HAMLET is a protein-lipid complex with a specific and broad bactericidal and tumoricidal activity, that lacks cytotoxic activity against healthy cells. In this study, we show that HAMLET also has general immune-stimulatory effects on primary human monocyte-derived dendritic cells and macrophages (Mo-DC and Mo-M) and murine RAW264.7 macrophages. HAMLET, but not its components alpha-lactalbumin or oleic acid, induces mature CD14low/- CD83+ Mo-DC and M1-like CD14+ CD86++ Mo-M surface phenotypes. Concomitantly, inflammatory mediators, including IL-2, IL-6, IL-10, IL-12 and MIP-1α, were released in the supernatant of HAMLET-stimulated cells, indicating a mainly pro-inflammatory phenotype. The HAMLET-induced phenotype was mediated by calcium, NFκB and p38 MAPK signaling in Mo-DCs and calcium, NFκB and ERK signaling in Mo-M as inhibitors of these pathways almost completely blocked the induction of mature Mo-DCs and M1-like Mo-M. Compared to unstimulated Mo-DCs, HAMLET-stimulated Mo-DCs were more potent in inducing T cell proliferation and HAMLET-stimulated macrophages were more efficient in phagocytosis of Streptococcus pneumoniae in vitro. This indicates a functionally activated phenotype of HAMLET-stimulated DCs and macrophages. Combined, we propose that HAMLET has a two-fold anti-bacterial activity; one inducing direct cytotoxic activity, the other indirectly mediating elimination of bacteria by activation of immune cells of the myeloid lineage.


Subject(s)
Cytokines/immunology , Inflammation Mediators/immunology , Lactalbumin/immunology , Myeloid Cells/immunology , Oleic Acids/immunology , Animals , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Inflammation Mediators/metabolism , Lactalbumin/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Myeloid Cells/drug effects , Myeloid Cells/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Oleic Acids/pharmacology , Phagocytosis/drug effects , Phagocytosis/immunology , Phenotype , RAW 264.7 Cells , Signal Transduction/drug effects , Signal Transduction/immunology , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
17.
ACS Chem Neurosci ; 11(24): 4416-4425, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33253528

ABSTRACT

Parkinson's Disease (PD) is characterized by increased oxidative stress and decreased level of dopamine. At present, the therapeutic interventions of PD are associated with undesirable adverse effects. To overcome these side effects, a new candidate bioinspired molecule is needed for the management of PD. Camel α-lactalbumin (α-LA) is the most abundant protein in camel's milk and has a potential to act as a nutraceutical supplement for neurological functions. Oleic acid, a monounsaturated fatty acid, has been widely associated with a reduced risk of PD. The present study aimed to formulate the camel α-LA and oleic acid (CLOA) complex under specific conditions and to evaluate its efficacy as a neuroprotective in rotenone induced PC12 cell model of PD. Our results demonstrated that removal of Ca++ ions from camel α-LA by EDTA enhances its binding efficiency with oleic acid, and the complex was characterized by UV-CD, ANS fluorescence spectroscopy, and NMR spectroscopy. Moreover, CLOA complex treatment reduced the oxidative stress and increased the cell viability by enhancing the level of dopamine and the expression of SIRT1, FOXO3a, HIF-1α, and HSF-1. We also validated the neuroprotective role of the complex by incubating the cells with CLOA complex prior to rotenone treatment. We inferred from the outcome of the results that the individual entity, i.e., α-LA or OA, is not as effective as the complex. Taken together, our study indicates that CLOA complex might be a potential candidate for the development of future therapeutic drugs for PD.


Subject(s)
Lactalbumin , Parkinson Disease , Animals , Camelus , Lactalbumin/pharmacology , Oleic Acid/pharmacology , Sirtuin 1
18.
Int J Biol Macromol ; 165(Pt A): 205-213, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-32991904

ABSTRACT

The antioxidant and antibacterial activities of camel and bovine α-lactalbumin (α-La) in both calcium-loaded (holo) and calcium-depleted (apo) forms were investigated and compared. Antioxidant assay showed that camel and bovine α-La exhibited significant Ferric-reducing antioxidant power (FRAP), ferrous iron-chelating activity (FCA) and antiradical activities especially in their apo form. Camel apo α-La also exhibited attractive antibacterial activities against Gram-negative bacteria (Pseudomonas aeruginosa) and against fungal pathogens species (Penicillium bilaiae, Aspergillus tamari and Aspergillus sclerotiorum). Likewise, emulsifying properties (emulsification ability (EAI) and stability (ESI) indexes) and the surface characteristics (surface hydrophobicity, ζ-potential and interfacial tension) of the α-La were assessed. Maximum EAI were found at pH 7.0, with higher EAI values for the camel apo α-La (EAI ~19.5 m2/g). This behavior was explained by its relative high surface hydrophobicity and its greater efficiency to reduce the surface tension at the oil-water interface. Furthermore, emulsions were found to be more stable at pH 7.0 compared to pH 5.0 (ESI ~50%) due to the higher electrostatic repulsive forces between oil droplets at pH 7.0 in consistence with the ζ-potential results. This study concluded that the camel apo α-La has antibacterial, antioxidant, and emulsifying properties in agricultural and food industries.


Subject(s)
Anti-Bacterial Agents/chemistry , Antioxidants/chemistry , Lactalbumin/chemistry , Animals , Anti-Bacterial Agents/pharmacology , Antioxidants/isolation & purification , Antioxidants/pharmacology , Apoenzymes/chemistry , Apoenzymes/isolation & purification , Aspergillus/drug effects , Camelus , Cattle , Emulsions/chemistry , Emulsions/pharmacology , Holoenzymes/chemistry , Holoenzymes/isolation & purification , Hydrophobic and Hydrophilic Interactions/drug effects , Lactalbumin/isolation & purification , Lactalbumin/pharmacology , Penicillium/drug effects
19.
Int J Biol Macromol ; 163: 1697-1706, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32961181

ABSTRACT

We investigated whether the modification of the negatively charged carboxyl groups with semicarbazide could confer membrane-disrupting and cytotoxic properties to bovine α-lactalbumin (LA). MALDI-TOF analysis revealed that eighteen of the twenty-one carboxyl groups in LA were coupled with semicarbazide molecules. Measurement of circular dichroism spectra and Trp fluorescence quenching studies showed that semicarbazide-modified LA (SEM-LA) had a molten globule-like conformation that retained the α-helix secondary structure but lost the tertiary structure of LA. Compared to LA, SEM-LA had a higher structural flexibility in response to trifluoroethanol- and temperature-induced structural transitions. In sharp contrast to LA, SEM-LA exhibited membrane-damaging activity and cytotoxicity. Furthermore, SEM-LA-induced membrane permeability promoted the uptake of daunorubicin and thereby its cytotoxicity. The microenvironment surrounding the Trp residues of SEM-LA was enriched in positive charges, as revealed by iodide quenching studies. The binding of SEM-LA with lipid vesicles altered the positively charged cluster around Trp residues. Although LA and SEM-LA displayed similar lipid-binding affinities, the membrane interaction modes of SEM-LA and LA differed. Collectively, these results suggest that blocking of negatively charged residues enables the formation of a molten-globule conformation of LA with structural flexibility and increased positive charge, thereby generating functional LA with membrane-disrupting activity and cytotoxicity.


Subject(s)
Cell Membrane/drug effects , Cytotoxins/metabolism , Cytotoxins/pharmacology , Lactalbumin/metabolism , Lactalbumin/pharmacology , Animals , Cattle , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Circular Dichroism , Humans , Protein Structure, Secondary , Protein Structure, Tertiary , Trifluoroethanol/metabolism , Trifluoroethanol/pharmacology , U937 Cells
20.
Molecules ; 25(13)2020 Jun 30.
Article in English | MEDLINE | ID: mdl-32630113

ABSTRACT

Identifying DPP-IV inhibitory peptides from dietary protein has attracted increased attention. In the present study, bovine α-lactalbumin hydrolysates were generated by alcalase for various hydrolysis times, and DPP-IV inhibitory activity of these hydrolysates was determined. The 4 h hydrolysates displayed the most potent DPP-IV inhibitory activity, with DPP-IV inhibition rate of 82.30 ± 1.39% at concentration of 1.0 mg/mL. DPP-IV inhibitory peptides were isolated from the 4 h-hydrolysates with gel filtration chromatography and reversed-phase high-performance liquid chromatography (RP-HPLC). Using liquid chromatography-electrospray ionization tandem mass spectrometry (LC-ESI MS/MS), two DPP-IV inhibitory peptides were identified, and their amino acid sequences were Glu-Leu-Lys-Asp-Leu-Lys-Gly-Tyr (ELKDLKGY) and Ile-Leu-Asp-Lys-Val-Gly-Ile-Asn-Tyr (ILDKVGINY), respectively. Furthermore, molecular docking analysis showed that peptides ELKDLKGY and ILDKVGINY could form hydrogen bonds, pi-cation interactions, and salt bridges with DPP-IV. These findings indicated that bovine α-lactalbumin may be a potential source of natural DPP-IV inhibitor.


Subject(s)
Dipeptidyl Peptidase 4/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Lactalbumin/pharmacology , Peptide Fragments/pharmacology , Animals , Cattle , Dipeptidyl Peptidase 4/metabolism , Peptide Fragments/classification
SELECTION OF CITATIONS
SEARCH DETAIL