Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (41)

Search Parameters:
Keywords = adenine diet model

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 9552 KiB  
Article
Benefits of Maternal Choline Supplementation on Aged Basal Forebrain Cholinergic Neurons (BFCNs) in a Mouse Model of Down Syndrome and Alzheimer’s Disease
by Melissa J. Alldred, Harshitha Pidikiti, Kyrillos W. Ibrahim, Sang Han Lee, Adriana Heguy, Gabriela Chiosis, Elliott J. Mufson, Grace E. Stutzmann and Stephen D. Ginsberg
Biomolecules 2025, 15(8), 1131; https://doi.org/10.3390/biom15081131 - 5 Aug 2025
Abstract
Down syndrome (DS), stemming from the triplication of human chromosome 21, results in intellectual disability, with early mid-life onset of Alzheimer’s disease (AD) pathology. Early interventions to reduce cognitive impairments and neuropathology are lacking. One modality, maternal choline supplementation (MCS), has shown beneficial [...] Read more.
Down syndrome (DS), stemming from the triplication of human chromosome 21, results in intellectual disability, with early mid-life onset of Alzheimer’s disease (AD) pathology. Early interventions to reduce cognitive impairments and neuropathology are lacking. One modality, maternal choline supplementation (MCS), has shown beneficial effects on behavior and gene expression in neurodevelopmental and neurodegenerative disorders, including trisomic mice. Loss of basal forebrain cholinergic neurons (BFCNs) and other DS/AD relevant hallmarks were observed in a well-established trisomic model (Ts65Dn, Ts). MCS attenuates these endophenotypes with beneficial behavioral effects in trisomic offspring. We postulate MCS ameliorates dysregulated cellular mechanisms within vulnerable BFCNs, with attenuation driven by novel gene expression. Here, choline acetyltransferase immunohistochemical labeling identified BFCNs in the medial septal/ventral diagonal band nuclei of the basal forebrain in Ts and normal disomic (2N) offspring at ~11 months of age from dams exposed to MCS or normal choline during the perinatal period. BFCNs (~500 per mouse) were microisolated and processed for RNA-sequencing. Bioinformatic assessment elucidated differentially expressed genes (DEGs) and pathway alterations in the context of genotype (Ts, 2N) and maternal diet (MCS, normal choline). MCS attenuated select dysregulated DEGs and relevant pathways in aged BFCNs. Trisomic MCS-responsive improvements included pathways such as cognitive impairment and nicotinamide adenine dinucleotide signaling, among others, indicative of increased behavioral and bioenergetic fitness. Although MCS does not eliminate the DS/AD phenotype, early choline delivery provides long-lasting benefits to aged trisomic BFCNs, indicating that MCS prolongs neuronal health in the context of DS/AD. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

11 pages, 1895 KiB  
Article
Comparison of Adenine-Induced Rat Models for Vascular Calcification in Chronic Kidney Disease
by Ho Won Kang, Ji Hye Kim, A Ro Yoon, Jahyung Kim, Joonhee Kim, Min Gyu Kyung and Dong Yeon Lee
Biology 2025, 14(7), 814; https://doi.org/10.3390/biology14070814 - 4 Jul 2025
Viewed by 347
Abstract
Vascular calcification significantly contributes to cardiovascular complications and limb loss in chronic kidney disease (CKD). To establish an optimal rat model for vascular calcification, we tested varying adenine concentrations and feeding durations in Sprague–Dawley rats (n = 72), divided into six groups (n [...] Read more.
Vascular calcification significantly contributes to cardiovascular complications and limb loss in chronic kidney disease (CKD). To establish an optimal rat model for vascular calcification, we tested varying adenine concentrations and feeding durations in Sprague–Dawley rats (n = 72), divided into six groups (n = 12 each). The control group received a standard diet for 18 weeks. Group 1 was given 0.5% adenine for 4 weeks, followed by a standard diet. Group 2 received 0.5% adenine for 4 weeks, then 0.25% for 14 weeks. Group 3A received 0.5% adenine for 12 weeks and then standard diet; group 3B received 0.5% adenine for 12 weeks, followed by 0.25% for 6 weeks; group 3C received 0.5% adenine for 18 weeks. At week 18, vascular calcification was absent in the control and group 1. Groups 2 and 3A showed low incidence (12.5%), while groups 3B and 3C showed high incidence (66.7%). However, survival rates differed: 75.0% in 3B and 50.0% in 3C after 12 wk. Thus, 0.5% adenine for 12 weeks followed by 0.25% for 6 weeks effectively induced vascular calcification while maintaining acceptable survival, providing a practical model for studying CKD-related vascular pathology. Full article
(This article belongs to the Section Medical Biology)
Show Figures

Figure 1

13 pages, 2763 KiB  
Communication
Comparison of Anti-Renal Fibrosis Activity of Eucommiae cortex Extract and Its Microbial Fermentation Products
by Zhengyou He, Wenyi Jiang, Ruijiao Yao, Wenyan Xiao, Zhiyang Chen, Miao Zheng, Xia Zeng, Jia Li, Zhengwen Li and Yong Jiang
Pharmaceuticals 2025, 18(5), 747; https://doi.org/10.3390/ph18050747 - 19 May 2025
Viewed by 501
Abstract
Background: Renal fibrosis is a common pathological feature of all progressive chronic kidney disease (CKD). Eucommiae cortex (EC) is a valuable economic tree species endemic to China. The microbial fermentation of Chinese medicines can release their active ingredients as effectively as possible or [...] Read more.
Background: Renal fibrosis is a common pathological feature of all progressive chronic kidney disease (CKD). Eucommiae cortex (EC) is a valuable economic tree species endemic to China. The microbial fermentation of Chinese medicines can release their active ingredients as effectively as possible or produce new active ingredients with enhanced efficacy and reduced toxic side effects; Methods: The microbial fermentation of EC can produce pinoresinol (Pin) and dehydrodiconiferyl alcohol (DA). In this study, C57 BL/6 mice were fed a diet containing 0.2% adenine, resulting in a model of chronic kidney disease. The effects of EC and EC ferment (ECF) on CKD were explored by the exogenous supplementation of EC and ECF; Results: The results of the study showed that exogenous supplementation with EC and ECF suc-cessfully reduced creatinine and urea nitrogen levels, down-regulated the expression levels of TGF-β1, α-SMA, Smad3, and phospho-Smad3 in the TGF-β1/Smad signaling pathway, and ameliorated renal fibrosis; Conclusions: Both EC and ECF may have reno-protective effects and provide a reference for relevant clinical drug development. Full article
(This article belongs to the Section Natural Products)
Show Figures

Graphical abstract

22 pages, 2923 KiB  
Article
Crosstalk Among Gut Microbiota, Fecal Metabolites, and Amygdala Neuropathology Genes After Ginger Polyphenol Administration in Female Rats with Neuropathic Pain: Evidence for Microbiota–Gut–Brain Connection
by Chwan-Li Shen, Julianna Maria Santos, Moamen M. Elmassry, Fang Chen, Guangchen Ji, Peyton Presto, Takaki Kiritoshi, Xiaobo Liu and Volker Neugebauer
Nutrients 2025, 17(9), 1444; https://doi.org/10.3390/nu17091444 - 25 Apr 2025
Viewed by 879
Abstract
Objectives. The relationships among neuropathic pain, gut microbiota, microbiome-derived metabolites, and neuropathology have received increasing attention. This study examined the effects of two dosages of gingerol-enriched ginger (GEG) on mechanical hypersensitivity, anxiety-like behavior, gut microbiome composition and its metabolites, and neuropathology markers in [...] Read more.
Objectives. The relationships among neuropathic pain, gut microbiota, microbiome-derived metabolites, and neuropathology have received increasing attention. This study examined the effects of two dosages of gingerol-enriched ginger (GEG) on mechanical hypersensitivity, anxiety-like behavior, gut microbiome composition and its metabolites, and neuropathology markers in female rats in the spinal nerve ligation (SNL) model of neuropathic pain. Methods. Forty female rats were assigned to 4 groups: sham-vehicle, SNL-vehicle, SNL+GEG at 200 mg/kg BW, and SNL+GEG at 600 mg/kg BW via oral gavage. All animals were given an AIN-93G diet for 5 weeks. Mechanical hypersensitivity was assessed by the von Frey test. Anxiety-like behavior was assessed by the open field test. Fecal microbiota composition and metabolites were determined using 16S rRNA gene sequencing and GC-MS, respectively. Neuropathology gene expression profiling of the amygdala was assessed by an nCounter® Neuropathology pathway panel. Results. Both GEG-treated groups showed decreased mechanical hypersensitivity and anxiety-like behavior in the SNL model. Gut microbiome diversity in both GEG groups was decreased compared with untreated SNL rats. In the SNL model, phyla such as Bacteroidota, Proteobacteria and Verrucomicrobiota were decreased. Compared with the untreated SNL group, both GEG groups exhibited increased abundance of the phyla Bacteroidota (i.e., Rikenella, Alistipes, Muribaculaceae, Odoribacter), Firmicutes (i.e., UBA1819, Ruminococcaceae, Oscillospiraceae, Roseburia), and Verrucomicrobiota (i.e., Victivallis). GEG groups had higher levels of nine hydrophilic positive metabolites [val-glu, urocanic acid, oxazolidinone, L-threonine, L-norleucine, indole, imino-tryptophan, 2,3-octadiene-5,7-diyn-1-ol, and (2E)-3-(3-hydroxyphenyl) acrylaldehyde] and two hydrophilic negative metabolites [methylmalonic acid and metaphosphoric acid], as well as lower levels of five hydrophilic metabolites [xanthine, N-acetylmuramic acid, doxaprost, adenine, and 1-myristoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine]. Among the 770 neuropathology genes, 1 gene (PLA2G4A) was upregulated and 2 genes (CDK5R1 and SHH) were downregulated in SNL rats. GEG caused the upregulation of nine genes (APC, CCNH, EFNA5, GRN, HEXB, ITPR1, PCSK2, TAF9, and WFS1) and downregulation of three genes (AVP, C4A, and TSPO) in the amygdala. Conclusions. GEG supplementation mitigated pain-associated behaviors in female rats with neuropathic pain, in part by reversing the molecular neuropathology signature of the amygdala. This was associated with changes in the gut microbiome composition and fecal metabolites, which could play a role in mediating the effects of GEG on neuropathic pain. Full article
(This article belongs to the Section Nutrition and Public Health)
Show Figures

Figure 1

19 pages, 3137 KiB  
Article
Inhibiting Myostatin Expression by the Antisense Oligonucleotides Improves Muscle Wasting in a Chronic Kidney Disease Mouse Model
by Arju Akhter, Abdullah Md. Sheikh, Jun Yoshino, Takeshi Kanda, Atsushi Nagai, Masafumi Matsuo and Shozo Yano
Int. J. Mol. Sci. 2025, 26(7), 3098; https://doi.org/10.3390/ijms26073098 - 27 Mar 2025
Viewed by 2524
Abstract
Sarcopenia, a serious consequence of chronic kidney disease (CKD), is driven by elevated myostatin (MSTN), a key inhibitor of muscle growth. This study explored the potential of an MSTN-specific antisense oligonucleotide (ASO) in reversing CKD-induced muscle wasting in a mouse model. Thirty-two male [...] Read more.
Sarcopenia, a serious consequence of chronic kidney disease (CKD), is driven by elevated myostatin (MSTN), a key inhibitor of muscle growth. This study explored the potential of an MSTN-specific antisense oligonucleotide (ASO) in reversing CKD-induced muscle wasting in a mouse model. Thirty-two male C57BL/6J mice were randomly assigned to a non-CKD group (n = 8, regular diet) and a CKD group (n = 24, adenine diet). CKD was induced using a 0.2% adenine-supplemented diet for 4 weeks. Following this, the mice were sub-grouped into CKD (saline, n = 8), CKD + Low-Dose ASO (25 mg/kg ASO, n = 8), and CKD + High-Dose ASO (50 mg/kg ASO, n = 8). ASO was administered via subcutaneous injections for 8 weeks. Muscle mass, treadmill performance, grip strength, and muscle fiber morphology were assessed alongside qPCR and Western blot analysis for MSTN, atrogin-1, and MuRF-1 expression. ASO therapy significantly enhanced muscle mass and function and enlarged muscle fibers while effectively downregulating muscle degradation markers. These improvements occurred without compromising renal function, as confirmed by BUN, creatinine, kidney weight, and histological analysis. This study is the first to demonstrate the efficacy of ASO therapy in mitigating CKD-induced sarcopenia, offering a promising targeted gene therapy with significant clinical implications for improving nutritional status and physical performance in CKD. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapies in Skeletal Muscle Diseases)
Show Figures

Figure 1

15 pages, 998 KiB  
Review
Beneficial Effects of Butyrate on Kidney Disease
by Tram N. Diep, Haoxin Liu and Liang-Jun Yan
Nutrients 2025, 17(5), 772; https://doi.org/10.3390/nu17050772 - 22 Feb 2025
Cited by 2 | Viewed by 2045
Abstract
The gut microbiota influences and contributes to kidney health and disease. Butyrate, a short-chain fatty acid molecule generated via the fermentation of gut bacterial catabolism of nondigestible dietary fiber, has been shown to exert numerous beneficial effects on kidney disorders. The objective of [...] Read more.
The gut microbiota influences and contributes to kidney health and disease. Butyrate, a short-chain fatty acid molecule generated via the fermentation of gut bacterial catabolism of nondigestible dietary fiber, has been shown to exert numerous beneficial effects on kidney disorders. The objective of this review was to discuss the latest findings on the protective effects of butyrate on a variety of animal models of kidney injury. We conducted a PubMed search using the title word “butyrate” and keyword “kidney” to generate our literature review sources. The animal models covered in this review include ischemia–reperfusion renal injury, cisplatin- and folic acid-induced kidney injury, septic kidney injury, diabetic kidney disease (DKD), high-fat diet (HFD)-induced glomerulopathy, adenine-induced chronic kidney disease (CKD), high-salt-induced renal injury, and T-2 toxin-induced kidney injury in birds. The protective mechanisms of butyrate that are most shared among these animal model studies include antioxidative stress, anti-fibrosis, anti-inflammation, and anti-cell death. This review ends with suggestions for future studies on potential approaches that may modulate gut microbiota butyrate production for the well-being of kidneys with the kidney disorders covered in this review. Full article
Show Figures

Figure 1

13 pages, 2587 KiB  
Article
A Simplified Model of Adenine-Induced Chronic Kidney Disease Using SKH1 Mice
by Benjamin W. French, Joshua D. Breidenbach, Shereen G. Yassine, Bella Z. Khatib-Shahidi, Sara Kazmi, Caitlin M. Murphy, Humza S. Bashir, Evan M. Benson, Bivek Timalsina, Upasana Shrestha, Dhilhani Faleel, Satkeerth Boyapalli, Prabhatchandra Dube, Apurva Lad, Irum Syed, Deepak Malhotra, Amira Gohara, David J. Kennedy and Steven T. Haller
Cells 2024, 13(24), 2117; https://doi.org/10.3390/cells13242117 - 20 Dec 2024
Cited by 1 | Viewed by 2504
Abstract
Commonly used adenine-induced chronic kidney disease (CKD) murine models often employ C57BL/6 mice; however, this strain has inherent limitations due to its natural resistance to developing key pathological features of CKD, such as tubulointerstitial fibrosis and inflammation. There have been attempts to overcome [...] Read more.
Commonly used adenine-induced chronic kidney disease (CKD) murine models often employ C57BL/6 mice; however, this strain has inherent limitations due to its natural resistance to developing key pathological features of CKD, such as tubulointerstitial fibrosis and inflammation. There have been attempts to overcome these barriers by using multiple concentrations of adenine-supplemented diets or by performing prolonged experiments up to 20 weeks. Here, we demonstrate that SKH1 Elite mice develop clinically relevant CKD phenotypes (e.g., polyuria, proteinuria, inflammation, and renal fibrosis) over the course of only 6 weeks of low-dose (0.15%) adenine supplementation. As a docile, immunocompetent, and hairless strain, SKH1 Elite mice offer several logistical advantages over C57BL/6 mice, including ease of handling and the ability to study dermal conditions, which are often secondary to CKD. Full article
(This article belongs to the Special Issue Cellular and Molecular Basis in Chronic Kidney Disease)
Show Figures

Figure 1

12 pages, 1833 KiB  
Article
Antihypertensive Effects of a Sodium Thiosulfate-Loaded Nanoparticle in a Juvenile Chronic Kidney Disease Rat Model
by You-Lin Tain, Chien-Ning Hsu, Chih-Yao Hou and Chih-Kuang Chen
Antioxidants 2024, 13(12), 1574; https://doi.org/10.3390/antiox13121574 - 20 Dec 2024
Cited by 1 | Viewed by 1082
Abstract
Sodium thiosulfate (STS), a precursor of hydrogen sulfide (H2S), has demonstrated antihypertensive properties. Previous studies have suggested that H2S-based interventions can prevent hypertension in pediatric chronic kidney disease (CKD). However, the clinical application of STS is limited by its [...] Read more.
Sodium thiosulfate (STS), a precursor of hydrogen sulfide (H2S), has demonstrated antihypertensive properties. Previous studies have suggested that H2S-based interventions can prevent hypertension in pediatric chronic kidney disease (CKD). However, the clinical application of STS is limited by its rapid release and intravenous administration. To address this, we developed a poly-lactic acid (PLA)-based nanoparticle system for sustained STS delivery and investigated whether weekly treatment with STS-loaded nanoparticles (NPs) could protect against hypertension in a juvenile CKD rat model. Male Sprague Dawley rats, aged three weeks, were fed a diet containing 0.5% adenine for three weeks to induce a model of pediatric CKD. STS-loaded NPs (25 mg/kg) were administered intravenously during weeks 6, 7, and 8, and at week 9, all rats were sacrificed. Treatment with STS-loaded NPs reduced systolic and diastolic blood pressure by 10 mm Hg and 8 mm Hg, respectively, in juvenile CKD rats. The protective effect of STS-loaded NPs was linked to increased renal expression of H2S-producing enzymes, including cystathionine γ-lyase (CSE) and D-amino acid oxidase (DAO). Additionally, STS-loaded NP therapy restored nitric oxide (NO) signaling, increasing L-arginine levels, which were disrupted in CKD. Furthermore, the beneficial effects of STS-loaded NPs were associated with inhibition of the renin–angiotensin system (RAS) and the enhancement of the NO signaling pathway. Our findings suggest that STS-loaded NP treatment provides sustained STS delivery and effectively reduces hypertension in a juvenile CKD rat model, bringing us closer to the clinical translation of STS-based therapy for pediatric CKD-induced hypertension. Full article
Show Figures

Figure 1

24 pages, 4268 KiB  
Article
Sex-Specific Changes in Cardiac Function and Electrophysiology During Progression of Adenine-Induced Chronic Kidney Disease in Mice
by Valentina Dargam, Anet Sanchez, Aashiya Kolengaden, Yency Perez, Rebekah Arias, Ana M. Valentin Cabrera, Daniel Chaparro, Christopher Tarafa, Alexandra Coba, Nathan Yapaolo, Perony da Silva Nogueira, Emily A. Todd, Monique M. Williams, Lina A. Shehadeh and Joshua D. Hutcheson
J. Cardiovasc. Dev. Dis. 2024, 11(11), 362; https://doi.org/10.3390/jcdd11110362 - 7 Nov 2024
Cited by 2 | Viewed by 1637
Abstract
Chronic kidney disease (CKD) and cardiovascular disease (CVD) often co-exist, with notable sex-dependent differences in manifestation and progression despite both sexes sharing similar risk factors. Identifying sex-specific diagnostic markers in CKD-induced CVD could elucidate why the development and progression of these diseases differ [...] Read more.
Chronic kidney disease (CKD) and cardiovascular disease (CVD) often co-exist, with notable sex-dependent differences in manifestation and progression despite both sexes sharing similar risk factors. Identifying sex-specific diagnostic markers in CKD-induced CVD could elucidate why the development and progression of these diseases differ by sex. Adult, C57BL/6J male and female mice were fed a high-adenine diet for 12 weeks to induce CKD, while control mice were given a normal diet. Adenine-treated males showed more severe CKD than females. Cardiac physiology was evaluated using electrocardiogram (ECG) and echocardiogram markers. Only adenine-treated male mice showed markers of left ventricular (LV) hypertrophy. Adenine males showed markers of LV systolic and diastolic dysfunction throughout regimen duration, worsening as the disease progressed. Adenine males had prolonged QTc interval compared to adenine females and control males. We identified a new ECG marker, Speak-J duration, which increased with disease progression and appeared earlier in adenine-treated males than in females. We identified sex-dependent differences in cardiac structure, function, and electrophysiology in a CKD-induced CVD mouse model, with adenine-treated males displaying markers of LV hypertrophy, dysfunction, and electrophysiological changes. This study demonstrates the feasibility of using this model to investigate sex-dependent cardiac differences resulting from CKD. Full article
(This article belongs to the Section Electrophysiology and Cardiovascular Physiology)
Show Figures

Figure 1

11 pages, 3820 KiB  
Article
Changes in Skeletal Muscle Atrophy over Time in a Rat Model of Adenine-Induced Chronic Kidney Disease
by Kento Okamoto, Yuji Kasukawa, Koji Nozaka, Hiroyuki Tsuchie, Daisuke Kudo, Hayato Kinoshita, Yuichi Ono, Shun Igarashi, Fumihito Kasama, Shuntaro Harata, Keita Oya, Takashi Kawaragi, Kenta Tominaga, Manabu Watanabe and Naohisa Miyakoshi
Appl. Sci. 2024, 14(19), 9106; https://doi.org/10.3390/app14199106 - 9 Oct 2024
Cited by 1 | Viewed by 1523
Abstract
This study evaluated changes over time in skeletal muscle atrophy, expressions of skeletal muscle anabolic and catabolic genes, and mitochondrial activity by skeletal muscle type in an adenine-induced chronic kidney disease (CKD) model. A CKD model was successfully established by feeding male Wistar [...] Read more.
This study evaluated changes over time in skeletal muscle atrophy, expressions of skeletal muscle anabolic and catabolic genes, and mitochondrial activity by skeletal muscle type in an adenine-induced chronic kidney disease (CKD) model. A CKD model was successfully established by feeding male Wistar rats a 0.75% adenine diet for 4 weeks starting at 8 weeks of age. Control and CKD groups were sacrificed at 12 and 20 weeks of age. The back muscles were analyzed histologically, and succinate dehydrogenase (SDH) staining was performed to evaluate mitochondrial activity. Gene expressions of myogenic determination gene number 1 and myogenin as indicators of muscle anabolism, atrogin-1 and muscle RING-finger protein-1 (MuRF1) as indicators of muscle catabolism, and peroxisome proliferator-activated receptor-γ coactivator-1-α as a marker of mitochondrial biogenesis were assessed. Type I and type II muscle cross-sectional areas (CSAs) were decreased at 12 weeks, but type I muscle CSA was recovered at 20 weeks. SDH staining was lower in CKD than in control rats at 12 weeks, but no significant difference was observed at 20 weeks. Increased expressions of myogenin, atrogin-1, and MuRF-1 were observed only at 12 weeks, but no differences were observed at 20 weeks. The adenine-induced CKD rat model appears to show changes in muscle atrophy over time. Full article
Show Figures

Figure 1

19 pages, 5105 KiB  
Article
Hyperphosphatemia Contributes to Skeletal Muscle Atrophy in Mice
by Kylie Heitman, Seth Bollenbecker, Jordan Bradley, Brian Czaya, Abul Fajol, Sarah Madison Thomas, Qing Li, Svetlana Komarova, Stefanie Krick, Glenn C. Rowe, Matthew S. Alexander and Christian Faul
Int. J. Mol. Sci. 2024, 25(17), 9308; https://doi.org/10.3390/ijms25179308 - 28 Aug 2024
Cited by 2 | Viewed by 2490
Abstract
Chronic kidney disease (CKD) is associated with various pathologic changes, including elevations in serum phosphate levels (hyperphosphatemia), vascular calcification, and skeletal muscle atrophy. Elevated phosphate can damage vascular smooth muscle cells and cause vascular calcification. Here, we determined whether high phosphate can also [...] Read more.
Chronic kidney disease (CKD) is associated with various pathologic changes, including elevations in serum phosphate levels (hyperphosphatemia), vascular calcification, and skeletal muscle atrophy. Elevated phosphate can damage vascular smooth muscle cells and cause vascular calcification. Here, we determined whether high phosphate can also affect skeletal muscle cells and whether hyperphosphatemia, in the context of CKD or by itself, is associated with skeletal muscle atrophy. As models of hyperphosphatemia with CKD, we studied mice receiving an adenine-rich diet for 14 weeks and mice with deletion of Collagen 4a3 (Col4a3−/−). As models of hyperphosphatemia without CKD, we analyzed mice receiving a high-phosphate diet for three and six months as well as a genetic model for klotho deficiency (kl/kl). We found that adenine, Col4a3−/−, and kl/kl mice have reduced skeletal muscle mass and function and develop atrophy. Mice on a high-phosphate diet for six months also had lower skeletal muscle mass and function but no significant signs of atrophy, indicating less severe damage compared with the other three models. To determine the potential direct actions of phosphate on skeletal muscle, we cultured primary mouse myotubes in high phosphate concentrations, and we detected the induction of atrophy. We conclude that in experimental mouse models, hyperphosphatemia is sufficient to induce skeletal muscle atrophy and that, among various other factors, elevated phosphate levels might contribute to skeletal muscle injury in CKD. Full article
(This article belongs to the Special Issue Chronic Kidney Disease: The State of the Art and Future Perspectives)
Show Figures

Figure 1

14 pages, 3416 KiB  
Article
Lactoferrin Supplementation during Pregnancy and Lactation Protects Adult Male Rat Offspring from Hypertension Induced by Maternal Adenine Diet
by You-Lin Tain, Chih-Yao Hou, Wei-Ling Chen, Wei-Ting Liao and Chien-Ning Hsu
Nutrients 2024, 16(16), 2607; https://doi.org/10.3390/nu16162607 - 8 Aug 2024
Cited by 2 | Viewed by 2288
Abstract
Lactoferrin, a glycoprotein derived from breastmilk, is recognized for its health benefits in infants and children; however, its protective effects when administered during gestation and lactation against offspring hypertension remain unclear. This study aimed to investigate whether maternal lactoferrin supplementation could prevent hypertension [...] Read more.
Lactoferrin, a glycoprotein derived from breastmilk, is recognized for its health benefits in infants and children; however, its protective effects when administered during gestation and lactation against offspring hypertension remain unclear. This study aimed to investigate whether maternal lactoferrin supplementation could prevent hypertension in offspring born to mothers with chronic kidney disease (CKD), with a focus on nitric oxide (NO), renin–angiotensin system (RAS) regulation, and alterations in gut microbiota and short-chain fatty acids (SCFAs). Prior to pregnancy, female rats were subjected to a 0.5% adenine diet for 3 weeks to induce CKD. During pregnancy and lactation, pregnant rats received one of four diets: normal chow, 0.5% adenine diet, 10% lactoferrin diet, or adenine diet supplemented with lactoferrin. Male offspring were euthanized at 12 weeks of age (n = 8 per group). Supplementation with lactoferrin during gestation and lactation prevented hypertension in adult offspring induced by a maternal adenine diet. The maternal adenine diet caused a decrease in the index of NO availability, which was restored by 67% with maternal LF supplementation. Additionally, LF was related to the regulation of the RAS, as evidenced by a reduced renal expression of renin and the angiotensin II type 1 receptor. Combined maternal adenine and LF diets altered beta diversity, shifted the offspring’s gut microbiota, decreased propionate levels, and reduced the renal expression of SCFA receptors. The beneficial effects of lactoferrin are likely mediated through enhanced NO availability, rebalancing the RAS, and alterations in gut microbiota composition and SCFAs. Our findings suggest that maternal lactoferrin supplementation improves hypertension in offspring in a model of adenine-induced CKD, bringing us closer to potentially translating lactoferrin supplementation clinically for children born to mothers with CKD. Full article
(This article belongs to the Special Issue Breastmilk for Healthy Development)
Show Figures

Figure 1

20 pages, 2258 KiB  
Article
Alcohol- and Low-Iron Induced Changes in Antioxidant and Energy Metabolism Associated with Protein Lys Acetylation
by Jesse A. Thornton, Zeynep C. Koc, Vincent E. Sollars, Monica A. Valentovic, James Denvir, John Wilkinson and Emine C. Koc
Int. J. Mol. Sci. 2024, 25(15), 8344; https://doi.org/10.3390/ijms25158344 - 30 Jul 2024
Cited by 1 | Viewed by 1849
Abstract
Understanding the role of iron in ethanol-derived hepatic stress could help elucidate the efficacy of dietary or clinical interventions designed to minimize liver damage from chronic alcohol consumption. We hypothesized that normal levels of iron are involved in ethanol-derived liver damage and reduced [...] Read more.
Understanding the role of iron in ethanol-derived hepatic stress could help elucidate the efficacy of dietary or clinical interventions designed to minimize liver damage from chronic alcohol consumption. We hypothesized that normal levels of iron are involved in ethanol-derived liver damage and reduced dietary iron intake would lower the damage caused by ethanol. We used a pair-fed mouse model utilizing basal Lieber-DeCarli liquid diets for 22 weeks to test this hypothesis. In our mouse model, chronic ethanol exposure led to mild hepatic stress possibly characteristic of early-stage alcoholic liver disease, seen as increases in liver-to-body weight ratios. Dietary iron restriction caused a slight decrease in non-heme iron and ferritin (FeRL) expression while it increased transferrin receptor 1 (TfR1) expression without changing ferroportin 1 (FPN1) expression. It also elevated protein lysine acetylation to a more significant level than in ethanol-fed mice under normal dietary iron conditions. Interestingly, iron restriction led to an additional reduction in nicotinamide adenine dinucleotide (NAD+) and NADH levels. Consistent with this observation, the major mitochondrial NAD+-dependent deacetylase, NAD-dependent deacetylase sirtuin-3 (SIRT3), expression was significantly reduced causing increased protein lysine acetylation in ethanol-fed mice at normal and low-iron conditions. In addition, the detection of superoxide dismutase 1 and 2 levels (SOD1 and SOD2) and oxidative phosphorylation (OXPHOS) complex activities allowed us to evaluate the changes in antioxidant and energy metabolism regulated by ethanol consumption at normal and low-iron conditions. We observed that the ethanol-fed mice had mild liver damage associated with reduced energy and antioxidant metabolism. On the other hand, iron restriction may exacerbate certain activities of ethanol further, such as increased protein lysine acetylation and reduced antioxidant metabolism. This metabolic change may prove a barrier to the effectiveness of dietary reduction of iron intake as a preventative measure in chronic alcohol consumption. Full article
Show Figures

Figure 1

21 pages, 3862 KiB  
Article
Identification and Evaluation of Olive Phenolics in the Context of Amine Oxidase Enzyme Inhibition and Depression: In Silico Modelling and In Vitro Validation
by Tom C. Karagiannis, Katherine Ververis, Julia J. Liang, Eleni Pitsillou, Siyao Liu, Sarah M. Bresnehan, Vivian Xu, Stevano J. Wijoyo, Xiaofei Duan, Ken Ng, Andrew Hung, Erik Goebel and Assam El-Osta
Molecules 2024, 29(11), 2446; https://doi.org/10.3390/molecules29112446 - 23 May 2024
Cited by 3 | Viewed by 2576
Abstract
The Mediterranean diet well known for its beneficial health effects, including mood enhancement, is characterised by the relatively high consumption of extra virgin olive oil (EVOO), which is rich in bioactive phenolic compounds. Over 200 phenolic compounds have been associated with Olea europaea [...] Read more.
The Mediterranean diet well known for its beneficial health effects, including mood enhancement, is characterised by the relatively high consumption of extra virgin olive oil (EVOO), which is rich in bioactive phenolic compounds. Over 200 phenolic compounds have been associated with Olea europaea, and of these, only a relatively small fraction have been characterised. Utilising the OliveNetTM library, phenolic compounds were investigated as potential inhibitors of the epigenetic modifier lysine-specific demethylase 1 (LSD1). Furthermore, the compounds were screened for inhibition of the structurally similar monoamine oxidases (MAOs) which are directly implicated in the pathophysiology of depression. Molecular docking highlighted that olive phenolics interact with the active site of LSD1 and MAOs. Protein–peptide docking was also performed to evaluate the interaction of the histone H3 peptide with LSD1, in the presence of ligands bound to the substrate-binding cavity. To validate the in silico studies, the inhibitory activity of phenolic compounds was compared to the clinically approved inhibitor tranylcypromine. Our findings indicate that olive phenolics inhibit LSD1 and the MAOs in vitro. Using a cell culture model system with corticosteroid-stimulated human BJ fibroblast cells, the results demonstrate the attenuation of dexamethasone- and hydrocortisone-induced MAO activity by phenolic compounds. The findings were further corroborated using human embryonic stem cell (hESC)-derived neurons stimulated with all-trans retinoic acid. Overall, the results indicate the inhibition of flavin adenine dinucleotide (FAD)-dependent amine oxidases by olive phenolics. More generally, our findings further support at least a partial mechanism accounting for the antidepressant effects associated with EVOO and the Mediterranean diet. Full article
Show Figures

Figure 1

12 pages, 2975 KiB  
Article
Cognitive Impairment Related to Chronic Kidney Disease Is Associated with a Decreased Abundance of Membrane-Bound Klotho in the Cerebral Cortex
by María E. Rodríguez-Ortiz, Daniel Jurado-Montoya, Karen Valdés-Díaz, Raquel M. García-Sáez, Ana I. Torralbo, Teresa Obrero, Victoria Vidal-Jiménez, María J. Jiménez, Andrés Carmona, Fátima Guerrero, María V. Pendón-Ruiz de Mier, Cristian Rodelo-Haad, Antonio Canalejo, Mariano Rodríguez, Sagrario Soriano-Cabrera and Juan R. Muñoz-Castañeda
Int. J. Mol. Sci. 2024, 25(8), 4194; https://doi.org/10.3390/ijms25084194 - 10 Apr 2024
Cited by 1 | Viewed by 1842
Abstract
Cognitive impairment (CI) is a complication of chronic kidney disease (CKD) that is frequently observed among patients. The aim of this study was to evaluate the potential crosstalk between changes in cognitive function and the levels of Klotho in the brain cortex in [...] Read more.
Cognitive impairment (CI) is a complication of chronic kidney disease (CKD) that is frequently observed among patients. The aim of this study was to evaluate the potential crosstalk between changes in cognitive function and the levels of Klotho in the brain cortex in an experimental model of CKD. To induce renal damage, Wistar rats received a diet containing 0.25% adenine for six weeks, while the control group was fed a standard diet. The animals underwent different tests for the assessment of cognitive function. At sacrifice, changes in the parameters of mineral metabolism and the expression of Klotho in the kidney and frontal cortex were evaluated. The animals with CKD exhibited impaired behavior in the cognitive tests in comparison with the rats with normal renal function. At sacrifice, CKD-associated mineral disorder was confirmed by the presence of the expected disturbances in the plasma phosphorus, PTH, and both intact and c-terminal FGF23, along with a reduced abundance of renal Klotho. Interestingly, a marked and significant decrease in Klotho was observed in the cerebral cortex of the animals with renal dysfunction. In sum, the loss in cerebral Klotho observed in experimental CKD may contribute to the cognitive dysfunction frequently observed among patients. Although further studies are required, Klotho might have a relevant role in the development of CKD-associated CI and represent a potential target in the management of this complication. Full article
(This article belongs to the Special Issue New Insights into CKD and Age-Related Bone and Mineral Disorders)
Show Figures

Figure 1

Back to TopTop