Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 281
1.
Eur J Pharm Sci ; 167: 106030, 2021 Dec 01.
Article En | MEDLINE | ID: mdl-34601071

Somapacitan is a reversible albumin-binding growth hormone (GH) derivative in clinical development for once-weekly administration in patients with adult GH deficiency (AGHD) and children with GH deficiency (GHD). To date, the use of somapacitan in AGHD or severe AGHD has been approved in the USA and Japan, respectively. This study (ClinicalTrials.gov, NCT02962440) investigated the absorption, metabolism and excretion, as well as the pharmacokinetics (PK), of tritium-labelled somapacitan ([3H]-somapacitan). Seven healthy males received a single subcutaneous dose of 6 mg somapacitan containing [3H]-somapacitan 20 MBq. Blood, serum, plasma, urine, faeces, and expired air were collected for radioactivity assessment. Metabolites were identified and quantified in plasma and urine collected. The PK of plasma components were determined, and the radioactive peaks of the most abundant plasma metabolites and urine metabolites were selected for analysis. Twenty-eight days after dosing, 94.0% of the administered dose was recovered as [3H]-somapacitan-related material, most of which was excreted in urine (80.9%); 12.9% was excreted in faeces, and an insignificant amount (0.2%) was exhaled in expired air. PK properties of [3H]-somapacitan-related material appeared to be consistent across plasma, serum and blood. Three abundant plasma metabolites (P1, M1 and M1B) and two abundant urine metabolites (M4 and M5) were identified. The total exposure of intact somapacitan accounted for 59% of the total exposure of all somapacitan-related material, P1 accounted for 21% and M1 plus M1B accounted for 12%. M4 and M5 were the most abundant urine metabolites and accounted for 37% and 8% of the dosed [3H]-somapacitan radioactivity, respectively. No intact somapacitan was found in excreta. Two subjects had six adverse events (AEs); all were mild in severity and unlikely to be related to trial product. The majority of dosed [3H]-somapacitan (94%) was recovered as excreted metabolites. Urine was the major route for excretion of somapacitan metabolites, followed by faeces, and exhalation in expired air was negligible. The low molecular weights of identified urine metabolites demonstrate that somapacitan was extensively degraded to small residual fragments that were excreted (fully biodegradable). The extensive metabolic degradation and full elimination of metabolites in excreta were the major clearance pathways of somapacitan and the key elements in its biological fate. A single dose of 6 mg somapacitan (containing [3H]-somapacitan) in healthy male subjects was well tolerated with no unexpected safety issues identified.


Histidine/administration & dosage , Histidine/pharmacokinetics , Human Growth Hormone/administration & dosage , Human Growth Hormone/pharmacokinetics , Mannitol/administration & dosage , Mannitol/pharmacokinetics , Phenol/administration & dosage , Phenol/pharmacokinetics , Administration, Cutaneous , Administration, Oral , Adult , Albumins , Child , Feces , Histidine/urine , Human Growth Hormone/urine , Humans , Male , Mannitol/urine , Phenol/urine , Research Subjects
2.
J Trace Elem Med Biol ; 65: 126723, 2021 May.
Article En | MEDLINE | ID: mdl-33508549

BACKGROUND: The objective of this study was to investigate the effects of different chromium histidinate (CrHis) complexes added to the diet of rats fed a high-fat diet (HFD) on body weight changes, glucose and lipid metabolism parameters, and changes in biomarkers such as PPAR-γ, IRS-1, GLUTs, and NF-κB proteins. METHODS: Forty-two Sprague-Dawley rats were divided equally into six groups and fed either a control, an HFD, or an HFD supplemented with either CrHis1, CrHis2, CrHis3, or a combination of the CrHis complexes as CrHisM. RESULTS: Feeding an HFD to rats increased body weights, HOMA-IR values, fasting serum glucose, insulin, leptin, free fatty acid, total cholesterol, low-density lipoprotein cholesterol, and MDA concentrations as well as AST activities, and decreased serum and brain serotonin concentrations compared with rats fed a control diet (P < 0.0001). The levels of the PPAR-γ, IRS-1, and GLUTs in the liver and brain decreased, while NF-κB level increased, with feeding an HFD (P < 0.05). Although all the CrHis supplements reversed the negative effects of feeding an HFD (P < 0.05), the CrHis1 complex was most effective in changing the protein levels, while CrHisM was most effective in influencing certain parameters such as body weight and serum metabolites. CONCLUSION: The results of the present work suggest that the CrHis1 complex is most potent for alleviating the negative effects of feeding an HFD. The efficacy of CrHisM is likely due to the presence of the CrHis1 complex.


Blood Glucose/drug effects , Glucose Transporter Type 1/antagonists & inhibitors , Histidine/analogs & derivatives , Insulin Receptor Substrate Proteins/antagonists & inhibitors , NF-kappa B/metabolism , Organometallic Compounds/pharmacology , PPAR gamma/antagonists & inhibitors , Animals , Diet, High-Fat/adverse effects , Dietary Supplements , Glucose Transporter Type 1/metabolism , Histidine/administration & dosage , Histidine/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Lipid Metabolism/drug effects , Organometallic Compounds/administration & dosage , PPAR gamma/metabolism , Rats , Rats, Sprague-Dawley
3.
J Dairy Sci ; 104(2): 1759-1776, 2021 Feb.
Article En | MEDLINE | ID: mdl-33453803

Two His deletion studies were conducted to examine the mechanisms used by dairy cows to support milk true protein yield (MTPY) when His supply is altered. The potential mechanisms involved in how the efficiency of utilization of His varied included reduced catabolism, more efficient mammary usage, and use of His labile pools. For the first study, 5 multicatheterized cows were used in a 4 × 4 Latin square plus 1 cow with 14-d periods. Treatments were abomasal infusion of increasing doses of His (0, 7.6, 15.2, and 20.8 g/d) in addition to a mixture of AA (595 g/d; casein profile excluding His). Cows were fed the same protein-deficient diet throughout the study. The MTPY increased linearly with a quadratic tendency with increasing doses of His. Muscle concentrations of carnosine, a His-based dipeptide, tended to increase in a quadratic manner with increasing His supply, suggesting that the 0- and 7.6-g doses were insufficient to cover His requirement. Liver catabolism of His decreased as His supply decreased. Mammary fractional removal of His was considerably greater at low His supply, but the ratio of His mammary net uptake to milk output was not affected by the rate of His infusion, averaging 1.02. The mechanisms to face a reduced His supply included reduced His hepatic catabolism, more efficient His mammary use of lowered arterial supply, and, to a lesser extent, use of His labile pools. Two independent estimates of His efficiency were calculated, one based on the sum of exported proteins (measured MTPY plus estimated metabolic fecal protein and scurf; i.e., the anabolic component, EffMTPY) and the other based on liver removal (i.e., the catabolic component). These 2 estimates followed the same pattern of response to His supply, decreasing with increasing His supply. The EffMTPY at which MTPY peaked was 0.785. For the second study, 6 cows were used in a 6 × 6 Latin square with 7-d periods. Two greater doses of His (30.4 and 38.0 g/d) were added; otherwise, the nutritional design was similar to the first study. In this second study, the indicator AA oxidation technique was used instead of the multiorgan approach, with labeled Leu as the indicator of His utilization. The MTPY peaked and Leu oxidation reached the nadir at an average EffMTPY of 0.763. Combined across both studies, the data indicate that optimal usage of His would occur at a threshold EffMTPY of 0.77. The agreement between experimental approaches across both studies indicates that the biological optimal supply of His expressed in grams per day could be calculated as the sum of exported proteins divided by this EffMTPY plus estimated endogenous urinary excretion.


Cattle/metabolism , Histidine/metabolism , Lactation/physiology , Milk Proteins/metabolism , Milk/metabolism , Abomasum/drug effects , Abomasum/metabolism , Amino Acids/metabolism , Animals , Caseins/metabolism , Diet/veterinary , Female , Histidine/administration & dosage , Liver/metabolism , Mammary Glands, Animal/metabolism
4.
Mediators Inflamm ; 2020: 1240152, 2020.
Article En | MEDLINE | ID: mdl-33354159

Previous study showed that low protein diet-fed pigs are characterized by lower histidine concentration in the serum and muscle, suggesting that histidine may involve in protein-restricted response. Thus, the current study mainly investigated the effects of dietary histidine on growth performance, blood biochemical parameters and amino acids, intestinal morphology, and microbiota communities in low protein diet-challenged-piglets. The results showed that protein restriction inhibited growth performance, blood biochemical parameters and amino acids, and gut microbiota but had little effect on intestinal morphology. Dietary supplementation with histidine markedly enhanced serum histidine level and restored tryptophan concentration in low protein diet-fed piglets, while growth performance and intestinal morphology were not markedly altered in histidine-treated piglets. In addition, histidine exposure failed to affect bacterial diversity (observed species, Shannon, Simpson, Chao1, ACE, and phylogenetic diversity), but histidine-treated piglets exhibited higher abundances of Butyrivibrio and Bacteroides compared with the control and protein-restricted piglets. In conclusion, dietary histidine in low protein diet enhanced histidine concentration and affected gut microbiota (Butyrivibrio and Bacteroides) but failed to improve growth performance and intestinal morphology.


Amino Acids/blood , Diet, Protein-Restricted , Gastrointestinal Microbiome , Histidine/administration & dosage , Intestines/pathology , Swine/growth & development , Animals , Diet , Male , Organ Size
5.
J Nutr ; 150(Suppl 1): 2606S-2608S, 2020 10 01.
Article En | MEDLINE | ID: mdl-33000163

Based on research presented during the 10th Amino Acid Assessment Workshop, no observed adverse effect levels (NOAELs) for supplemental methionine at 46 mg/(kg·d) (∼3.2 g/d), for supplemental histidine at 8.0 g/d, and for supplemental lysine at 6.0 g/d have been proposed. These NOAELs are relevant to healthy adults and are applicable only to high-purity amino acids administered in fortified foods or dietary supplements. Because individuals are exposed to the above supplemental amino acids in the context of complex combinations of essential amino acids or individually in dietary supplements for various physiologic benefits, such as body fat reduction, skin conditioning, mental energy increase, or herpes simplex treatments, the above safety recommendations will make an important contribution to regulatory and nutritional practices.


Dietary Supplements , Food, Fortified , Histidine/administration & dosage , Lysine/administration & dosage , Methionine/administration & dosage , Histidine/adverse effects , Histidine/metabolism , Humans , Lysine/adverse effects , Lysine/metabolism , Methionine/adverse effects , Methionine/metabolism , Reference Values
6.
Am J Clin Nutr ; 112(5): 1358-1367, 2020 11 11.
Article En | MEDLINE | ID: mdl-32766885

BACKGROUND: Histidine is an essential amino acid with health benefits that may warrant histidine supplementation; however, the clinical safety of histidine intake above the average dietary intake (1.52-5.20 g/d) needs to be vetted. OBJECTIVES: We aimed to determine the tolerance to graded dosages of histidine in a healthy adult population. METHODS: Healthy adults aged 21-50 y completed graded dosages of histidine supplement (4, 8, and 12 g/d, Study 1) (n = 20 men and n = 20 women) and/or a 16-g/d dosage of histidine (Study 2, n = 21 men and n = 19 women); 27 participants (n = 12 men and n = 15 women) completed both studies. After study enrollment and baseline measures, participants consumed encapsulated histidine for 4 wk followed by a 3-wk recovery period. Primary outcomes included vitals, select biochemical analytes, anthropometry, serum zinc, and body composition (via DXA). RESULTS: No changes in vitals or body composition occurred with histidine supplementation in either study. Plasma histidine (measured in subjects who completed all dosages for Studies 1 and 2) was elevated at the 12- and 16-g/d dosages (compared with 0-8 g/d, P < 0.05) and blood urea nitrogen increased with dosage (P = 0.013) and time (P < 0.001) in Study 1 and with time in Study 2 (P < 0.001). In Study 1, mean ferritin concentrations were lower in 12 g/d (46.0 ng/mL; 95% CI: 34.8, 60.9 ng/mL) than in 4 g/d (51.6 ng/mL; 95% CI: 39.0, 68.4 ng/mL; P = 0.038). In Study 2, 16 g/d increased mean aspartate aminotransferase from baseline (19 U/L; 95% CI: 17, 22 U/L) to week 4 (24 U/L; 95% CI: 21, 27 U/L; P < 0.001) and mean serum zinc decreased from baseline (0.75 µg/dL; 95% CI: 0.71, 0.80 µg/dL) to week 4 (0.70 µg/dL; 95% CI: 0.66, 0.74 µg/dL; P = 0.011). CONCLUSIONS: Although values remained within the normal reference ranges for all analytes measured, in all dosages tested, the human no-observed adverse effect level was determined to be 8 g/d owing to changes in blood parameters at the 12-g/d dosage.This trial was registered at clinicaltrials.gov as NCT04142294.


Histidine/pharmacology , Adult , Blood Glucose/drug effects , C-Reactive Protein , Dietary Supplements , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Histidine/administration & dosage , Histidine/adverse effects , Humans , Male , Middle Aged , Young Adult
7.
J Dairy Sci ; 103(8): 7110-7123, 2020 Aug.
Article En | MEDLINE | ID: mdl-32505393

Hydrolyzed feather meal (HFM) is high in crude protein, most of which bypasses rumen degradation when fed to lactating dairy cows, allowing direct supply of AA to the small intestine. Compared with other feeds that are high in bypass protein, such as blood meal or heat-treated soybean meal, HFM is low in His and Lys. The objectives of this study were to determine the effects of supplementing rumen-protected (RP) Lys and His individually or in combination in a diet containing 5% HFM on milk production and composition as well as energy and N partitioning. Twelve multiparous Jersey cows (mean ± SD: 91 ± 18 d in milk) were used in a triplicated 4 × 4 Latin square with 4 periods of 28 d (24-d adaptation and 4-d collection). Throughout the experiment, all cows were fed the same TMR, with HFM included at 5% of diet DM. Cows were grouped by dry matter intake and milk yield, and cows within a group were randomly assigned to 1 of 4 treatments: no RP Lys or RP His; RP Lys only [70 g/d of Ajipro-L (24 g/d of digestible Lys), Ajinomoto Co. Inc., Tokyo, Japan]; RP His only [32 g/d of experimental product (7 g/d of digestible His), Balchem Corp., New Hampton, NY]; or both RP Lys and His. Plasma Lys concentration increased when RP Lys was supplemented without RP His (77.7 vs. 66.0 ± 4.69 µM) but decreased when RP Lys was supplemented with RP His (71.4 vs. 75.0 ± 4.69 µM). Plasma concentration of 3-methylhistidine decreased with RP Lys (3.19 vs. 3.40 ± 0.31 µM). With RP His, plasma concentration of His increased (21.8 vs. 18.7 ± 2.95 µM). For milk production and milk composition, no effects of Lys were observed. Supplementing RP His increased milk yield (22.5 vs. 21.6 ± 2.04 kg/d) and tended to increase milk protein yield (0.801 vs. 0.772 ± 0.051 kg/d). Across treatments, dry matter intake (18.5 ± 0.83 kg/d) and energy supply (32.2 ± 2.24 Mcal of net energy for lactation) were not different. Supplementing RP His did not affect N utilization; however, supplementing RP Lys increased N balance (25 vs. 16 ± 9 g/d). The lack of production responses to RP Lys suggests that Lys was not limiting or that the increase in Lys supply was not large enough to cause an increase in milk protein yield. However, increased N balance and decreased 3-methylhistidine with RP Lys suggest that increased Lys supply increased protein accretion and decreased protein mobilization. Furthermore, His may be a limiting AA in diets containing HFM.


Cattle/psychology , Dietary Supplements/analysis , Histidine/administration & dosage , Lysine/administration & dosage , Milk/metabolism , Nitrogen/metabolism , Animal Feed/analysis , Animals , Diet/veterinary , Eating , Feathers , Female , Histidine/blood , Lactation/drug effects , Lysine/blood , Methylhistidines/blood , Milk Proteins/metabolism , Random Allocation , Rumen/metabolism , Glycine max
8.
Arch Anim Nutr ; 74(3): 237-255, 2020 Jun.
Article En | MEDLINE | ID: mdl-32159388

To formulate low-protein diets for blue foxes with sufficient amounts of amino acids (AA), AA digestibility and AA requirements of the animals are crucial information. Therefore, a digestibility and nitrogen (N) balance trial was conducted with 20 blue foxes to determine the macronutrient and AA digestibility and N utilisation in low-protein diets supplemented with DL-methionine (Met) and L-histidine (His). In addition, plasma urea and plasma AA were measured. The diets were designated as P24 (control), P20, P20M, P16M and P16MH and contained energy from digestible crude protein (DCP) at 24%, 20% or 16% of total dietary metabolisable energy (ME). The 20% protein level was fed with or without Met and the 16% protein level was fed with Met and with or without His. The apparent total-tract digestibility (ATTD) of crude protein linearly decreased with decreasing dietary protein level. The ATTD of dry matter, organic matter and crude carbohydrates increased when wheat starch was added as a replacement for protein. The apparent ileal digestibility (AID) and ATTD methods were compared to determine the AA digestibility. The decreasing dietary protein supply decreased the ATTD of most of the AA: threonine, tryptophan (Trp), valine, alanine (Ala), aspartic acid (Asp), glutamic acid, glycine (Gly), proline (Pro), serine (Ser) and total AA. The AID of the AA was constant between diets. Diverging AA showed higher or lower digestibility when determined in the AID or ATTD methods. Isoleucine, lysine, Met, Ala and tyrosine showed higher levels of AID. Arginine, His, cysteine (Cys), Trp, Asp, Gly, Pro and Ser showed higher levels of ATTD, which may reflect the net loss of these AA in the large intestine. Met and His supplementation improved the ATTD and AID of the AA in question, respectively, but did not affect the other variables examined. N retention did not differ between diets and renal N excretion decreased with decreasing protein level; thus N utilisation improved. It was concluded that the protein supply and AA composition in low-protein diets with supplemented Met were adequate for adult blue foxes, since the lower protein supply improved N utilisation and did not affect N retention. However, His supplementation failed to reach the designed level and therefore showed no clear results.


Diet, Protein-Restricted/veterinary , Digestion , Foxes/physiology , Histidine/metabolism , Ileum/physiology , Nitrogen/metabolism , Racemethionine/metabolism , Animal Feed/analysis , Animal Nutritional Physiological Phenomena/drug effects , Animals , Dietary Supplements/analysis , Digestion/drug effects , Histidine/administration & dosage , Male , Racemethionine/administration & dosage , Random Allocation
10.
Physiol Res ; 69(1): 99-111, 2020 02 19.
Article En | MEDLINE | ID: mdl-31852202

Histidine (HIS) is investigated for therapy of various disorders and as a nutritional supplement to enhance muscle performance. We examined effects of HIS on amino acid and protein metabolism. Rats consumed HIS in a drinking water at a dose of 0.5 g/l (low HIS), 2 g/l (high HIS) or 0 g/l (control) for 4 weeks. At the end of the study, the animals were euthanized and blood plasma, liver, soleus (SOL), tibialis (TIB), and extensor digitorum longus (EDL) muscles analysed. HIS supplementation increased food intake, body weight and weights and protein contents of the liver and kidneys, but not muscles. In blood plasma there were increases in glucose, urea, and several amino acids, particularly alanine, proline, aspartate, and glutamate and in high HIS group, ammonia was increased. The main findings in the liver were decreased concentrations of methionine, aspartate, and glycine and increased alanine. In muscles of HIS-consuming animals increased alanine and glutamine. In high HIS group (in SOL and TIB) increased chymotrypsin-like activity of proteasome (indicates increased proteolysis); in SOL decreased anserine (beta-alanyl-N1-methylhistidine). We conclude that HIS supplementation increases ammonia production, alanine and glutamine synthesis in muscles, affects turnover of proteins and HIS-containing peptides, and increases requirements for glycine and methionine.


Amino Acids/metabolism , Histidine/administration & dosage , Animals , Dietary Supplements , Liver/metabolism , Male , Muscles/metabolism , Proteasome Endopeptidase Complex/metabolism , Random Allocation , Rats, Wistar
11.
Amino Acids ; 51(10-12): 1667-1680, 2019 Nov.
Article En | MEDLINE | ID: mdl-31712921

The unique capability of proton buffering is the rationale for using histidine (HIS) as a component of solutions for induction of cardiac arrest and myocardial protection in cardiac surgery. In humans, infusion of cardioplegic solution may increase blood plasma HIS from ~ 70 to ~ 21,000 µM. We examined the effects of a large intravenous dose of HIS on ammonia and amino acid concentrations and energy status of the body. Rats received 198 mM HIS intravenously (20 ml/kg) or vehicle. Samples of blood plasma, urine, liver, and soleus (SOL) and extensor digitorum longus (EDL) muscles were analysed at 2 or 24 h after treatment. At 2 h after HIS load, we found higher HIS concentration in all examined tissues, higher urea and ammonia concentrations in blood and urine, lower ATP content and higher AMP/ATP ratio in the liver and muscles, higher concentrations of almost all examined amino acids in urine, and lower glycine concentration in blood plasma, liver, and muscles when compared with controls. Changes in other amino acids were tissue dependent, markedly increased alanine and glutamate in the blood and the liver. At 24 h, the main findings were lower ATP concentrations in muscles, lower concentrations of branched-chain amino acids (BCAA; valine, leucine, and isoleucine) in blood plasma and muscles, and higher carnosine content in SOL when compared with controls. It is concluded that a load of large HIS dose results in increased ammonia levels and marked alterations in amino acid and energy metabolism. Pathogenesis is discussed in the article.


Adenine Nucleotides/metabolism , Amino Acids/metabolism , Ammonia/metabolism , Histidine/metabolism , Administration, Intravenous , Amino Acids/chemistry , Animals , Cardioplegic Solutions/chemistry , Carnosine/metabolism , Energy Metabolism , Histidine/administration & dosage , Histidine/analysis , Ketoglutaric Acids/metabolism , Male , Organ Specificity , Rats , Rats, Wistar , Tissue Distribution , Urea/metabolism
12.
Exp Brain Res ; 237(10): 2481-2493, 2019 Oct.
Article En | MEDLINE | ID: mdl-31321447

Kainic acid (KA)-induced seizures and other experimental models of epilepsy have been proven to be instrumental in identifying novel targets that could be responsible for human icto- and epileptogenesis. We have previously shown that the ablation of pharmacoresistant voltage-gated Ca2+ channels with Cav2.3 as central ion-conducting pore (R-type Ca2+ channel) reduces the sensitivity towards KA-induced epilepsy in mice. In vivo, Cav2.3 channels are thought to be under tight allosteric control by endogenous loosely bound trace metal cations (Zn2+ and Cu2+) that suppress channel gating via a high-affinity trace metal-binding site. Metal dyshomeostasis in the brain, which is a common feature of (KA-induced) seizures, could therefore alter the normal function of Cav2.3 channels and may shift hippocampal and neocortical signaling towards hyperexcitation. To investigate the role of loosely bound metal ions for KA-induced hyperexcitation in vivo, we examined the effects of manipulating brain trace metal homeostasis in mice. To this end, we developed a murine system for intracerebroventricular administration of trace metal ions and/or histidine (His), which can bind Zn2+ and Cu2+ and is involved in their transendothelial transport at the blood-brain barrier. Unexpectedly, our preliminary findings indicate that application of His alone but not in the presence of Zn2+ has substantial beneficial effects on the outcome of KA-induced epilepsy in mice. As such, our results emphasize previous findings on the complex, two-sided role of loosely bound metal ions with regard to neuronal excitation and degeneration under pathophysiological conditions.


Hippocampus/drug effects , Histidine/pharmacology , Ions/metabolism , Seizures/drug therapy , Animals , Disease Models, Animal , Histidine/administration & dosage , Kainic Acid/pharmacology , Mice, Inbred C57BL , Seizures/chemically induced , Signal Transduction/drug effects
13.
Sci Rep ; 9(1): 9398, 2019 06 28.
Article En | MEDLINE | ID: mdl-31253836

The bovine lactoferricin L6 (RRWQWR) has been previously identified as a novel cell-penetrating peptide (CPP) that is able to efficiently internalize into human cells. L6 interacts with quantum dots (QDs) noncovalently to generate stable L6/QD complexes that enter cells by endocytosis. In this study, we demonstrate a modified L6 (HL6; CHHHHHRRWQWRHHHHHC), in which short polyhistidine peptides are introduced into both flanks of L6, has enhanced cell-penetrating ability in human bronchoalveolar carcinoma A549 cells. The mechanism of cellular uptake of HL6/QD complexes is primarily direct membrane translocation rather than endocytosis. Dimethyl sulfoxide (DMSO), but not pyrenebutyrate (PB), ethanol, oleic acid, or 1,2-benzisothiazol-3(2 H)-one (BIT), slightly enhances HL6-mediated protein transduction efficiency. Neither HL6 nor HL6/QD complexes are cytotoxic to A549 or HeLa cells. These results indicate that HL6 could be a more efficient drug carrier than L6 for biomedical as well as biotechnological applications, and that the function of polyhistidine peptides is critical to CPP-mediated protein transduction.


Cell Membrane/drug effects , Cell Membrane/metabolism , Cell-Penetrating Peptides/metabolism , Histidine , Amino Acid Sequence , Cell Line, Tumor , Cell-Penetrating Peptides/chemistry , Drug Carriers/chemistry , Drug Delivery Systems , Histidine/administration & dosage , Histidine/chemistry , Humans
14.
Gac Med Mex ; 155(2): 191-195, 2019.
Article En | MEDLINE | ID: mdl-31056589

Menkes disease is a neurodegenerative and lethal pathology caused by gene mutations of the copper-transporting ATP-7A enzyme; it manifests itself by neurological symptoms and connective tissue changes of varying severity. Timely subcutaneous use of copper histidinate (Cu-His) is determinant for quality of life. We report the first experiences in Mexico on Cu-His synthesis and its safe use in 3 cases where hypocupremia and hypoceruloplasminemia were corroborated. With advice of the Hospital for Sick Children of Toronto Canada, we prepared a 500 µg/mL solution. In all three cases were 250 µg of Cu-His applied without relevant undesirable effects for 30 days. Serum copper (Cu, expressed in µg/L) and ceruloplasmin (Cp, in mg/dL) were determined: case 1, Cu days 0 and 30, 8 and 504 µg/L; Cp days 0 and 30, 4 and 10.75 mg/dL; case 2, Cu days 0 and 30, <50 and 502 µg/L; Cp days 0 and 30, 2 and 15 mg/dL; case 3, Cu days 0 and 30, 3 and 84.2 µg/L; Cp days 0 and 30, 4 and 10.7 mg/dL. In Mexico, it is possible to safely synthesize Cu-His and treat MD, which must be intentionally sought.


La enfermedad de Menkes es una patología neurodegenerativa y letal debida a mutaciones génicas de la enzima ATP-7A trasportadora de cobre; se manifiesta por síntomas neurológicos y alteraciones del tejido conectivo de severidad variable. El uso subcutáneo oportuno de histidinato de cobre (Cu-His) es determinante en la calidad de vida. Se reportan las primeras experiencias en México en la síntesis y uso seguro de Cu-His en tres casos en los que corroboramos hipocupremia e hipoceruloplasminemia. Bajo asesoramiento del Hospital for Sick Children, Toronto, Canadá, elaboramos una solución de 500 µg/mL. En los tres casos aplicamos 250 µg de Cu-His, sin efectos indeseables relevantes durante 30 días y observamos las siguientes determinaciones séricas de cobre (Cu en µg/L) y ceruloplasmina (Cp en mg/dL): caso 1, Cu días 0 y 30, 8 y 504 µg/L; Cp días 0 y 30, 4 y 10.75 mg/dL; caso 2, Cu días 0 y 30, < 50 y 502, µg/L; Cp días 0 y 30, 2 y 15 mg/dL; caso 3, Cu días 0 y 30, 3 y 84.2 µg/L; Cp días 0 y 30, 4 y 10.7 mg/dL. En México es posible la síntesis segura de Cu-His y tratar la enfermedad de Menkes, la cual debe ser intencionalmente buscada.


Drug Compounding/methods , Histidine/analogs & derivatives , Menkes Kinky Hair Syndrome/drug therapy , Organometallic Compounds/administration & dosage , Quality of Life , Child, Preschool , Copper/blood , Histidine/administration & dosage , Histidine/adverse effects , Humans , Infant , Mexico , Organometallic Compounds/adverse effects , Pharmaceutical Solutions
15.
AAPS PharmSciTech ; 20(5): 169, 2019 Apr 19.
Article En | MEDLINE | ID: mdl-31004249

The aim of the present study was to explore the therapeutic efficacy of microemulsion-based delivery of histidine-capped silver nanoparticles in eradicating Klebsiella pneumoniae-induced burn wound infection. The developed microemulsion was characterized on the basis of differential light scattering, phase separation, refractive index, and specific conductance. Emulgel was prepared and characterized on the basis of thixotropy, texture, differential scanning calorimetry, and release kinetics. Emulgel was further evaluated in skin irritation and in vivo studies, namely full-thickness K. pneumoniae-induced burn wound infection treatment via topical route. Efficacy of treatment was evaluated in terms of bacterial load, histopathology, wound contraction, and other infection markers. The developed emulgel provided significant in vivo antibacterial activity of histidine-capped silver nanoparticle preparations via topical route and resulted in reduction in bacterial load, wound contraction, and enhanced skin healing as well as decrement of inflammatory markers such as malondialdehyde, myeloperoxidase, and reactive nitrogen intermediate compared to untreated animals. The present study encourages the further employment of histidine-capped silver nanoparticles along with microemulsion-based drug delivery system in combating antibiotic-resistant topical infections.


Anti-Infective Agents, Local/administration & dosage , Anti-Infective Agents, Local/therapeutic use , Burns/complications , Histidine/administration & dosage , Histidine/therapeutic use , Klebsiella Infections/drug therapy , Klebsiella pneumoniae , Silver Compounds/administration & dosage , Silver Compounds/therapeutic use , Wound Infection/drug therapy , Administration, Topical , Animals , Drug Delivery Systems , Emulsions , Female , Gels , Klebsiella Infections/microbiology , Metal Nanoparticles , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Wound Infection/microbiology
16.
Gac. méd. Méx ; 155(2): 191-195, mar.-abr. 2019. tab, graf
Article Es | LILACS | ID: biblio-1286482

Resumen La enfermedad de Menkes es una patología neurodegenerativa y letal debida a mutaciones génicas de la enzima ATP-7A trasportadora de cobre; se manifiesta por síntomas neurológicos y alteraciones del tejido conectivo de severidad variable. El uso subcutáneo oportuno de histidinato de cobre (Cu-His) es determinante en la calidad de vida. Se reportan las primeras experiencias en México en la síntesis y uso seguro de Cu-His en tres casos en los que corroboramos hipocupremia e hipoceruloplasminemia. Bajo asesoramiento del Hospital for Sick Children, Toronto, Canadá, elaboramos una solución de 500 µg/mL. En los tres casos aplicamos 250 µg de Cu-His, sin efectos indeseables relevantes durante 30 días y observamos las siguientes determinaciones séricas de cobre (Cu en µg/L) y ceruloplasmina (Cp en mg/dL): caso 1, Cu días 0 y 30, 8 y 504 µg/L; Cp días 0 y 30, 4 y 10.75 mg/dL; caso 2, Cu días 0 y 30, < 50 y 502, µg/L; Cp días 0 y 30, 2 y 15 mg/dL; caso 3, Cu días 0 y 30, 3 y 84.2 µg/L; Cp días 0 y 30, 4 y 10.7 mg/dL. En México es posible la síntesis segura de Cu-His y tratar la enfermedad de Menkes, la cual debe ser intencionalmente buscada.


Abstract Menkes disease is a neurodegenerative and lethal pathology caused by gene mutations of the copper-transporting ATP-7A enzyme; it manifests itself by neurological symptoms and connective tissue changes of varying severity. Timely subcutaneous use of copper histidinate (Cu-His) is determinant for quality of life. We report the first experiences in Mexico on Cu-His synthesis and its safe use in 3 cases where hypocupremia and hypoceruloplasminemia were corroborated. With advice of the Hospital for Sick Children of Toronto Canada, we prepared a 500 µg/mL solution. In all three cases were 250 µg of Cu-His applied without relevant undesirable effects for 30 days. Serum copper (Cu, expressed in µg/L) and ceruloplasmin (Cp, in mg/dL) were determined: case 1, Cu days 0 and 30, 8 and 504 µg/L; Cp days 0 and 30, 4 and 10.75 mg/dL; case 2, Cu days 0 and 30, <50 and 502 µg/L; Cp days 0 and 30, 2 and 15 mg/dL; case 3, Cu days 0 and 30, 3 and 84.2 µg/L; Cp days 0 and 30, 4 and 10.7 mg/dL. In Mexico, it is possible to safely synthesize Cu-His and treat MD, which must be intentionally sought.


Humans , Infant , Child, Preschool , Organometallic Compounds/administration & dosage , Quality of Life , Drug Compounding/methods , Histidine/analogs & derivatives , Menkes Kinky Hair Syndrome/drug therapy , Organometallic Compounds/adverse effects , Copper/blood , Pharmaceutical Solutions , Histidine/administration & dosage , Histidine/adverse effects , Mexico
17.
Poult Sci ; 98(8): 3194-3203, 2019 Aug 01.
Article En | MEDLINE | ID: mdl-30753623

The objective of this study was to evaluate the effects of in ovo injection of histidine on hatch performance and post-hatch development by determining hatchability, hatching time, BW gain, carcass traits, and intestinal morphology in domestic pigeons (Columba livia). A completely randomized design (n = 3) with a L-histidine injection treatment (His group, 0.55 mg histidine dissolved in 200 µL 0.75% saline as 1% Conc compared to total histidine in the egg), a 0.75% saline injection treatment (SC group), and non-injection treatment (NC group) was used. Six squabs from each treatment were randomly sampled at day of hatch (DOH), day 7 (D7), and day 14 (D14) post hatch, respectively. Results showed that in ovo injection of histidine solution increased (P < 0.01) the hatchability in comparison with that of other groups. The hatching time of His group was earlier (P = 0.05) than that of the NC group. In ovo injection of histidine had no influence (P > 0.05) on BW gain and carcass traits, but had a significant effect on the organ index of pigeons. The index of pancreas on DOH, the indices of leg, gizzard, proventriculus, small intestine, and pancreas on D7, and the index of head on D14 were significantly increased (P < 0.05) by injection of histidine. Moreover, in ovo injection of histidine had significant effects on the weight indices of different intestinal segments and jejunal morphology in squabs. Ileum weight index on D7 from His group was significantly higher (P < 0.05) than that of other groups. Compared with the NC group, the jejunal crypt depth on DOH and D7 of His group decreased (P < 0.05), while jejunal villus area and villus crypt ratio on D7 of His group increased (P < 0.05). The results of this study indicate that in ovo injection of histidine may have beneficial effects on squabs' hatchability, intestinal development but have negligible effects on their growth performance during early post-hatch period.


Columbidae/growth & development , Histidine/pharmacology , Animals , Body Composition/drug effects , Body Weight/drug effects , Histidine/administration & dosage , Injections/veterinary , Intestines/drug effects , Intestines/growth & development , Ovum , Random Allocation
18.
EMBO J ; 38(7)2019 04 01.
Article En | MEDLINE | ID: mdl-30804004

Rewired metabolism of glutamine in cancer has been well documented, but less is known about other amino acids such as histidine. Here, we use Drosophila cancer models to show that decreasing the concentration of histidine in the diet strongly inhibits the growth of mutant clones induced by loss of Nerfin-1 or gain of Notch activity. In contrast, changes in dietary histidine have much less effect on the growth of wildtype neural stem cells and Prospero neural tumours. The reliance of tumours on dietary histidine and also on histidine decarboxylase (Hdc) depends upon their growth requirement for Myc. We demonstrate that Myc overexpression in nerfin-1 tumours is sufficient to switch their mode of growth from histidine/Hdc sensitive to resistant. This study suggests that perturbations in histidine metabolism selectively target neural tumours that grow via a dedifferentiation process involving large cell size increases driven by Myc.


Cell Dedifferentiation , Central Nervous System Neoplasms/pathology , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Histidine/administration & dosage , Neural Stem Cells/pathology , Transcription Factors/metabolism , Animals , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/metabolism , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Female , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Male , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Transcription Factors/genetics
19.
Int J Pharm ; 559: 58-67, 2019 Mar 25.
Article En | MEDLINE | ID: mdl-30654063

Linear Polyethylenimine (lPEI) is an efficient cationic polymer for transfecting cells, both in vitro and in vivo, but poses concerns regarding cytotoxicity. Histidinylated lPEI (His-lPEI) exhibits also high transfection efficiency but lower cytotoxicity than lPEI. For the first time, we tested polyfection efficiency of polyplexes comprising both lPEI and His-lPEI. A series of pDNA polyplexes was prepared with mixtures of lPEI and His-lPEI and the amount of each polymer within His-lPEI/lPEI polyplexes was determined by flow cytometry. We show that His-lPEI/lPEI polyplexes exhibit properties similar to lPEI polyplexes in terms of size, morphology, assembly with pDNA, and polyplex stability while His-lPEI/lPEI polyplexes exhibit properties similar to His-lPEI polyplexes in terms of buffering capacity. Compared to polyplexes consisting only of lPEI or His-lPEI, the transfection profile reveals that His-lPEI/lPEI polyplexes containing 30% to 57% lPEI strongly increase polyfection efficiency of NIH3T3 fibroblasts and murine, as well as human skeletal muscle cell lines without cytotoxicity. Importantly, improved transfection of human dystrophin deficient skeletal muscle cell lines was obtained. These results indicate that His-lPEI/lPEI polyplexes are an improved non-viral vector for efficient transfection of dystrophin deficient skeletal muscle cell lines that should be tested on animals.


DNA , Histidine/chemistry , Muscle Cells/metabolism , Muscle Fibers, Skeletal/metabolism , Polyethyleneimine/administration & dosage , Polyethyleneimine/chemistry , Animals , Cell Line , Cytotoxins/administration & dosage , Cytotoxins/chemistry , DNA/chemistry , Gene Transfer Techniques , Histidine/administration & dosage , Humans , Mice , NIH 3T3 Cells , Polymers/chemistry , Transfection/methods
20.
J Nutr Biochem ; 65: 115-127, 2019 03.
Article En | MEDLINE | ID: mdl-30685580

Moderate dietary protein restriction promotes hyperphagia and thermogenesis; however, little is known of whether these responses are due to restriction of the essential amino acids tryptophan and histidine. Here, we determined whether restriction of tryptophan and histidine alone recapitulate the effects of total amino acid restriction on energy balance, and whether the metabolic responses are age-dependent. We fed young (12 weeks old) and older (29 weeks old) diet-induced obese rats with one of four high-fat diets: control (CON, 100% amino acid requirement), total amino acid restriction (TAA, 67% amino acid restriction), tryptophan restriction (TRP, 67% tryptophan restriction) or histidine restriction (HIS, 67% histidine restriction) for 21 days. Energy balance, hormones, and key markers of hepatic nutrient sensing and brown adipose thermogenesis were measured. We found that TAA increased food intake in both young and older rats, with TRP, but not HIS, transiently simulating the hyperphagia. TAA promoted sympathetically mediated increase in energy expenditure in young rats partly through increased ß2-adrenergic and FGF21 signaling in brown fat; TRP partially emulated these responses. TRP and HIS transiently increased fat mass in young rats, and TAA promoted adiposity in older rats. TAA, TRP and HIS increased postprandial FGF21 concentrations in older rats. TAA induced age-dependent differential changes in markers of hepatic amino acid sensing; TRP and HIS partially mimicked these responses. Collectively, restriction of tryptophan, but not histidine, partially recapitulated the age-dependent metabolic effects of total amino acid restriction, in concert with distinct changes in hepatic amino acid sensing and signaling mechanisms.


Energy Metabolism/drug effects , Obesity/etiology , Tryptophan/administration & dosage , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Age Factors , Amino Acids/administration & dosage , Animals , Behavior, Animal/drug effects , Body Composition/drug effects , Diet, High-Fat/adverse effects , Eating/drug effects , Histidine/administration & dosage , Hormones/metabolism , Liver/drug effects , Liver/metabolism , Male , Obesity/diet therapy , Proteins/metabolism , Rats, Sprague-Dawley
...