[go: up one dir, main page]

 
 
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
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,151)

Search Parameters:
Keywords = small-animal model

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
26 pages, 1144 KiB  
Review
Cancer Stem Cells in Oral Squamous Cell Carcinoma: A Narrative Review on Experimental Characteristics and Methodological Challenges
by Surendra Kumar Acharya, Saptarsi Shai, Yee Fan Choon, Indrayadi Gunardi, Firstine Kelsi Hartanto, Kathreena Kadir, Ajoy Roychoudhury, Rahmi Amtha and Vui King Vincent-Chong
Biomedicines 2024, 12(9), 2111; https://doi.org/10.3390/biomedicines12092111 - 16 Sep 2024
Viewed by 282
Abstract
Cancer stem cells (CSCs) represent a subpopulation of cancer cells that are believed to initiate and drive cancer progression. In animal models, xenotransplanted CSCs have demonstrated the ability to produce tumors. Since their initial isolation in blood cancers, CSCs have been identified in [...] Read more.
Cancer stem cells (CSCs) represent a subpopulation of cancer cells that are believed to initiate and drive cancer progression. In animal models, xenotransplanted CSCs have demonstrated the ability to produce tumors. Since their initial isolation in blood cancers, CSCs have been identified in various solid human cancers, including oral squamous cell carcinoma (OSCC). In addition to their tumorigenic properties, dysregulated stem-cell-related signaling pathways—Wnt family member (Wnt), neurogenic locus notch homolog protein (Notch), and hedgehog—have been shown to endow CSCs with characteristics like self-renewal, phenotypic plasticity, and chemoresistance, contributing to recurrence and treatment failure. Consequently, CSCs have become targets for new therapeutic agents, with some currently in different phases of clinical trials. Notably, small molecule inhibitors of the hedgehog signaling pathway, such as vismodegib and glasdegib, have been approved for the treatment of basal cell carcinoma and acute myeloid leukemia, respectively. Other strategies for eradicating CSCs include natural compounds, nano-drug delivery systems, targeting mitochondria and the CSC microenvironment, autophagy, hyperthermia, and immunotherapy. Despite the extensive documentation of CSCs in OSCC since its first demonstration in head and neck (HN) SCC in 2007, none of these novel pharmacological approaches have yet entered clinical trials for OSCC patients. This narrative review summarizes the in vivo and in vitro evidence of CSCs and CSC-related signaling pathways in OSCC, highlighting their role in promoting chemoresistance and immunotherapy resistance. Additionally, it addresses methodological challenges and discusses future research directions to improve experimental systems and advance CSC studies. Full article
(This article belongs to the Special Issue Novel Insights on Cancer Stem Cells)
Show Figures

Figure 1

Figure 1
<p>Evolution of cancer. (<b>A</b>) The cancer stem cell (CSC) theory of cancer evolution proposes the existence of a hierarchy in the heterogeneous cancer cell population in which cancer stem cells (CSCs) occupy the apex of the hierarchy. Through symmetrical and asymmetrical division in each cell cycle, CSCs give rise to other CSCs (through self-renewal) and non-CSCs, contributing to the heterogeneity in the cancer cell population. (<b>B</b>) In Nowell’s clonal theory of cancer evolution, all cancer cells are equally probable in acquiring more mutations, and each cell will expand clonally to form a heterogeneous cancer cell population.</p>
Full article ">Figure 2
<p>CSCs in OSCC. Cancer cells harvested from primary OSCC or cancer cell lines consist of a heterogeneous cancer population of CSCs and non-CSCs. CSCs are identified and then sorted by using cell surface markers. CD133 and CD44 are commonly used cell surface markers to identify and sort OSCC CSCs using fluorescence-assisted cell sorting (FASC) or magnetic beads in magnetic sorting for further characterization of CSCs. When cultured as a monolayer, CSCs grow as tightly packed cell colonies (holoclones). In non-adherent culture, CSCs readily form tumor spheres. Most importantly, CSCs form tumors rapidly when xenotransplanted into an animal model and resist chemotherapy regimens.</p>
Full article ">
15 pages, 8170 KiB  
Article
Assessment of Mannitol-Induced Chronic Blood–Brain Barrier Dysfunction In Vivo Using Magnetic Resonance
by Ana Sampedro-Viana, Sabela Fernández-Rodicio, José Castillo, Pablo Hervella, María Luz Alonso-Alonso and Ramón Iglesias-Rey
Int. J. Mol. Sci. 2024, 25(18), 9792; https://doi.org/10.3390/ijms25189792 - 10 Sep 2024
Viewed by 303
Abstract
The blood–brain barrier (BBB) is essential for protection and plays a crucial role in chronic neurological disorders like small-vessel disease and Alzheimer’s disease. Its complexity poses significant challenges for effective diagnostics and treatments, highlighting the need for novel animal models and comprehensive BBB [...] Read more.
The blood–brain barrier (BBB) is essential for protection and plays a crucial role in chronic neurological disorders like small-vessel disease and Alzheimer’s disease. Its complexity poses significant challenges for effective diagnostics and treatments, highlighting the need for novel animal models and comprehensive BBB dysfunction studies. This study investigates chronic BBB dysfunction induction using osmotic disruption via mannitol in healthy adult male Sprague Dawley rats over 12 weeks. Group 1 received 1 bolus/week (2.0 g/kg), Group 2 received 3 boluses/week (1.5 g/kg), and Group 3 received 3 boluses/week (2.5 g/kg). BBB dysfunction was assessed using gadolinium (Gd) infusion and MRI to evaluate location, severity, evolution, and persistence. MR spectroscopy (MRS) examined the brain metabolism changes due to intravenous mannitol, with T2-weighted MRI assessing brain lesions. Biomarkers of neuroinflammation were analyzed in the highest mannitol dose group. Our data show chronic BBB dysfunction primarily in the cortex, hippocampus, and striatum, but not in the corpus callosum of rats under periodic mannitol dosing in groups 1 and 2. MRS identified a distinctive metabolite signature, including changes in alanine, choline, and N-acetyl aspartate in the striatum of Group 1. No significant differences were found in the serum levels of all pro- and anti-inflammatory cytokines analyzed in the high-dose Group 3. This study underscores the feasibility and implications of using osmotic disruption to model chronic BBB dysfunction, offering insights for future neuroprotection and therapeutic strategies research. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Temporal changes in the animals’ weight. (<b>b</b>) Corpus callosum and striatum volumes between groups 1 and 3 at 12 weeks. (<b>c</b>) Changes in Evans blue extravasation in the entire brain and both hemispheres of rats at the end of the study (<span class="html-italic">n</span> = 3 per group). Extravasation of Evans blue was expressed as ng/mg brain tissue. The data are shown as mean ± SEM. The red line indicates normal levels. (<b>d</b>) Relative levels of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, and IL-2) anti-inflammatory cytokines (IL-10) in the mannitol-induced BBB dysfunction. Basal levels represent the measurements taken at week 0, where each analyte value was normalized by dividing it by its corresponding baseline. Values from the subsequent weeks were also normalized to each animal’s baseline. The data are described as the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>(<b>a</b>) Schematic representing the experimental protocol used for measuring the effects of repetitive mannitol i.v. injection on BBB dysfunction. In order to assess BBB permeability, magnetic resonance imaging with Gd contrast enhancement was performed at 0 (basal), 1, 3, and 12 weeks following the beginning of the study. Blood samples were collected at least 24 h after the last bolus at the same time to study inflammatory cytokines. MR spectroscopy was performed at identical time points in both groups 1 and 2, concluding with Evans blue staining. (<b>b</b>) Representative axial images of Gd-leakage in the brain at baseline (0 weeks), as well as at 6 and 12 weeks following the beginning of the study. Gd-leakage was calculated as the relative signal enhancement induced by Gd accumulation: the pre-contrast T1-weighted signal intensity was subtracted from the post-contrast signal intensity and divided by the pre-contrast signal intensity, as previously described [<a href="#B23-ijms-25-09792" class="html-bibr">23</a>,<a href="#B24-ijms-25-09792" class="html-bibr">24</a>]. The greater the number indicated by the color bar in the lower right corner, the closer the color on the image approaches white, indicating greater leakage. No significant Gd enhancement was detected in the brain before mannitol injections, except for the ventricles/circumventricular organs, which have an incomplete BBB. At 6 and 12 weeks, Gd-leakage was still detectable mainly in the hippocampus, striatum, and cortex. Arrows indicate higher extravasation values.</p>
Full article ">Figure 3
<p>Quantification of Gd-leakage at different follow-up points. T1-weighted MRI was performed to calculate the amount of Gd-leakage per region of interest (ROI) in the brain of the three groups studied. * <span class="html-italic">p</span> &lt; 0.05 , ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. groups, Kruskal–Wallis test followed by Mann–Whitney test.</p>
Full article ">Figure 4
<p>Metabolite differences between Group 1 (1 bolus/week (2.0 g/kg i.v., 25% solution)) and Group 2 (3 bolus/week (1.5 g/kg i.v., 25% solution)), * <span class="html-italic">p</span> &lt; 0.05. Metabolites: alanine (Ala), creatine (Cr), g-aminobutyric acid (GABA), glutamate (Glu), glutamine (Gln), glycine (Gly), N-acetylaspartate (NAA), phosphocholine (PCh or Ch), and taurine (Tau). Mannitol dosing periods correspond to the average metabolite levels observed in the subsequent weeks.</p>
Full article ">Figure 4 Cont.
<p>Metabolite differences between Group 1 (1 bolus/week (2.0 g/kg i.v., 25% solution)) and Group 2 (3 bolus/week (1.5 g/kg i.v., 25% solution)), * <span class="html-italic">p</span> &lt; 0.05. Metabolites: alanine (Ala), creatine (Cr), g-aminobutyric acid (GABA), glutamate (Glu), glutamine (Gln), glycine (Gly), N-acetylaspartate (NAA), phosphocholine (PCh or Ch), and taurine (Tau). Mannitol dosing periods correspond to the average metabolite levels observed in the subsequent weeks.</p>
Full article ">Figure 5
<p>Metabolite differences in Group 1 (1 bolus/week (2.0 g/kg i.v., 25% solution)), * <span class="html-italic">p</span> &lt; 0.05. Metabolites: alanine (Ala), creatine (Cr), g-aminobutyric acid (GABA), glutamate (Glu), glutamine (Gln), glycine (Gly), N-acetylaspartate (NAA), phosphocholine (PCh or Ch), and taurine (Tau). Basal levels represent the measurements taken at week 0, while the post-mannitol periods correspond to the average metabolite levels observed in the subsequent weeks.</p>
Full article ">Figure 5 Cont.
<p>Metabolite differences in Group 1 (1 bolus/week (2.0 g/kg i.v., 25% solution)), * <span class="html-italic">p</span> &lt; 0.05. Metabolites: alanine (Ala), creatine (Cr), g-aminobutyric acid (GABA), glutamate (Glu), glutamine (Gln), glycine (Gly), N-acetylaspartate (NAA), phosphocholine (PCh or Ch), and taurine (Tau). Basal levels represent the measurements taken at week 0, while the post-mannitol periods correspond to the average metabolite levels observed in the subsequent weeks.</p>
Full article ">
15 pages, 1195 KiB  
Review
Potential Therapeutic Interventions Targeting NAD+ Metabolism for ALS
by Samuel Lundt and Shinghua Ding
Cells 2024, 13(17), 1509; https://doi.org/10.3390/cells13171509 - 9 Sep 2024
Viewed by 652
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting both upper and lower motor neurons. While there have been many potential factors implicated for ALS development, such as oxidative stress and mitochondrial dysfunction, no exact mechanism has been determined at this time. [...] Read more.
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting both upper and lower motor neurons. While there have been many potential factors implicated for ALS development, such as oxidative stress and mitochondrial dysfunction, no exact mechanism has been determined at this time. Nicotinamide adenine dinucleotide (NAD+) is one of the most abundant metabolites in mammalian cells and is crucial for a broad range of cellular functions from DNA repair to energy homeostasis. NAD+ can be synthesized from three different intracellular pathways, but it is the NAD+ salvage pathway that generates the largest proportion of NAD+. Impaired NAD+ homeostasis has been connected to aging and neurodegenerative disease-related dysfunctions. In ALS mice, NAD+ homeostasis is potentially disrupted prior to the appearance of physical symptoms and is significantly reduced in the nervous system at the end stage. Treatments targeting NAD+ metabolism, either by administering NAD+ precursor metabolites or small molecules that alter NAD+-dependent enzyme activity, have shown strong beneficial effects in ALS disease models. Here, we review the therapeutic interventions targeting NAD+ metabolism for ALS and their effects on the most prominent pathological aspects of ALS in animal and cell models. Full article
(This article belongs to the Special Issue Genetics and Pathomechanisms of Amyotrophic Lateral Sclerosis (ALS))
Show Figures

Figure 1

Figure 1
<p>NAD<sup>+</sup> salvage pathway and NAD<sup>+</sup>-dependent enzymatic reactions. NAD<sup>+</sup> salvage pathway (<b>top</b>) NAM or NR are converted to NMN by NAMPT or NMRK, respectively. NMNAT generates NAD<sup>+</sup> from NMN. NAD<sup>+</sup> can be reversibly reduced and oxidized or utilized by NADases (SIRTs, PARPs, CD38, and SARM1), which produce NAM as a byproduct that can be re-used to form NAD<sup>+</sup>. NADase reactions (<b>bottom</b>). PARPs add ADPR to substrates. SIRTs remove acetyl groups from target substrates. CD38 and SARM1 generate ADPR/cADPR, which are important for second messenger signaling pathways. All figures were generated using BioRender.com.</p>
Full article ">Figure 2
<p>ALS pathophysiology and the effect of targeted interventions involving NAD<sup>+</sup> metabolism. Treating ALS models with therapeutic interventions altering NAD<sup>+</sup> metabolism ameliorates many disease-related impairments. Mitochondrial dysfunction, oxidative stress response, activation of glial cells, and protein mislocalization, all of which have been hypothesized as being involved in the development of ALS disease, are corrected from these interventions. Additionally, NMJ innervation and function, sites affected early during ALS development, are improved.</p>
Full article ">
14 pages, 2129 KiB  
Review
Exploring the Impact of Exercise-Derived Extracellular Vesicles in Cancer Biology
by Monica Silvestri, Elisa Grazioli, Guglielmo Duranti, Paolo Sgrò and Ivan Dimauro
Biology 2024, 13(9), 701; https://doi.org/10.3390/biology13090701 - 6 Sep 2024
Viewed by 852
Abstract
Cancer remains a major challenge in medicine, prompting exploration of innovative therapies. Recent studies suggest that exercise-derived extracellular vesicles (EVs) may offer potential anti-cancer benefits. These small, membrane-bound particles, including exosomes, carry bioactive molecules such as proteins and RNA that mediate intercellular communication. [...] Read more.
Cancer remains a major challenge in medicine, prompting exploration of innovative therapies. Recent studies suggest that exercise-derived extracellular vesicles (EVs) may offer potential anti-cancer benefits. These small, membrane-bound particles, including exosomes, carry bioactive molecules such as proteins and RNA that mediate intercellular communication. Exercise has been shown to increase EV secretion, influencing physiological processes like tissue repair, inflammation, and metabolism. Notably, preclinical studies have demonstrated that exercise-derived EVs can inhibit tumor growth, reduce metastasis, and enhance treatment response. For instance, in a study using animal models, exercise-derived EVs were shown to suppress tumor proliferation in breast and colon cancers. Another study reported that these EVs reduced metastatic potential by decreasing the migration and invasion of cancer cells. Additionally, exercise-induced EVs have been found to enhance the effectiveness of chemotherapy by sensitizing tumor cells to treatment. This review highlights the emerging role of exercise-derived circulating biomolecules, particularly EVs, in cancer biology. It discusses the mechanisms through which EVs impact cancer progression, the challenges in translating preclinical findings to clinical practice, and future research directions. Although research in this area is still limited, current findings suggest that EVs could play a crucial role in spreading molecules that promote better health in cancer patients. Understanding these EV profiles could lead to future therapies, such as exercise mimetics or targeted drugs, to treat cancer. Full article
(This article belongs to the Section Biochemistry and Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Exercise-conditioned serum inhibits signaling pathways that are crucial for cell proliferation. In cancer cells, increased proliferation is often driven by mutations in highly conserved signaling networks that regulate cell growth and division. When cancer cells are exposed to exercise-conditioned serum, several of these signaling pathways are altered, leading to a reduction in cell proliferation. For example, post-exercise serum has been shown to be enriched in catecholamines (e.g., norepinephrine and epinephrine), which can support the Hippo tumor suppressor pathway. When the Hippo pathway is “ON”, MST1/2 and MAP4K are activated, which subsequently phosphorylate and activate LATS1/2 kinases. Activated LATS1/2 phosphorylates transcriptional coactivator YAP/TAZ, preventing entry into the nucleus by promoting their degradation in the cytoplasm. Exercise has also been shown to increase the concentration of IGFBP1, a protein known to regulate cell proliferation, survival, differentiation, migration, and invasion. The binding of IGFBP1 with IGF1 modulates the activity of IGF1 signaling axes, such as the JAK, RAS, and AKT pathways, by regulating their availability to the IGF-IR. Similar effects were found with lower levels of EGF in exercise-conditioned serum, leading to reduced activation of the EGFR and its downstream signaling pathway. An antiproliferative effect appears to be derived from the increase in levels of some cytokines (i.e., TNFalpha, IL-6, OSM, and IL-8) induced by physical exercise. It is hypothesized that these molecules, in particular IL-6, released during exercise activate AMPK, which inhibits mTOR and its downstream effectors, such as p70s6k, as well as inhibits AKT and ERK1/2 phosphorylation/activation. Finally, the significant decrease in GSK3ß phosphorylation in exercise-conditioned serum highlights another possible antiproliferative effect of physical activity through the inhibition of the Wnt/ß-catenin pathway. MST1/2, Mammalian STE20-like 1/2; LATS1/2, Large Tumor Suppressor 1/2; YAP, Yes-associated protein; IGFBP1, insulin-like growth factor binding protein 1; IGF1, insulin-like growth factor; IGF-IR, IGF1 receptor; JAK, Janus tyrosine kinase; RAS; rat sarcoma virus; AKT, protein kinase B; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; TNFalpha, tumor necrosis factor alpha; IL-6, interleukine-6; OSM, oncostatin M; IL-8, interleukine-8.</p>
Full article ">Figure 2
<p>An overview depicting the impact of physical exercise on exercise-derived extracellular vesicles (EVs), emphasizing the molecular processes influenced by their cargo after exercise. EVs can be released from contracting skeletal muscles or other cell populations, particularly immune and endothelial cells, and then enter systemic circulation. Depending on their cargo (e.g., proteins, DNA, and ncRNAs), these EVs target various organs and affect several biological processes, including inflammation, immune response, cell survival, protein and nucleic acid metabolism, as well as stress and hormone responses. Notum, palmitoleoyl-protein carboxylesterase; Pctp, phosphatidylcholine transfer protein; Cyp4b1, cytochrome P450, family 4, subfamily b, polypeptide 1; Dnajb5, DnaJ Heat Shock Protein Family (Hsp40) Member B5; Hspa5, Heat Shock Protein Family A (Hsp70) Member 5; Ltb4r2, Leukotriene B4 receptor 2; Alox5, Arachidonate 5-lipoxygenase; Zbtb1, Zinc Finger And BTB Domain Containing 1; Oxtr, Oxytocin receptor; Dxo, Decapping exoribonuclease.</p>
Full article ">
31 pages, 4193 KiB  
Review
Realistic Aspects of Cardiac Ultrasound in Rats: Practical Tips for Improved Examination
by Jessica Silva, Tiago Azevedo, Mário Ginja, Paula A. Oliveira, José Alberto Duarte and Ana I. Faustino-Rocha
J. Imaging 2024, 10(9), 219; https://doi.org/10.3390/jimaging10090219 - 6 Sep 2024
Viewed by 446
Abstract
Echocardiography is a reliable and non-invasive method for assessing cardiac structure and function in both clinical and experimental settings, offering valuable insights into disease progression and treatment efficacy. The successful application of echocardiography in murine models of disease has enabled the evaluation of [...] Read more.
Echocardiography is a reliable and non-invasive method for assessing cardiac structure and function in both clinical and experimental settings, offering valuable insights into disease progression and treatment efficacy. The successful application of echocardiography in murine models of disease has enabled the evaluation of disease severity, drug testing, and continuous monitoring of cardiac function in these animals. However, there is insufficient standardization of echocardiographic measurements for smaller animals. This article aims to address this gap by providing a guide and practical tips for the appropriate acquisition and analysis of echocardiographic parameters in adult rats, which may also be applicable in other small rodents used for scientific purposes, like mice. With advancements in technology, such as ultrahigh-frequency ultrasonic transducers, echocardiography has become a highly sophisticated imaging modality, offering high temporal and spatial resolution imaging, thereby allowing for real-time monitoring of cardiac function throughout the lifespan of small animals. Moreover, it allows the assessment of cardiac complications associated with aging, cancer, diabetes, and obesity, as well as the monitoring of cardiotoxicity induced by therapeutic interventions in preclinical models, providing important information for translational research. Finally, this paper discusses the future directions of cardiac preclinical ultrasound, highlighting the need for continued standardization to advance research and improve clinical outcomes to facilitate early disease detection and the translation of findings into clinical practice. Full article
(This article belongs to the Section Medical Imaging)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the protocol for echocardiography in rodents. The probe positioning, the heart ultrasound image, and the parameters measurable in each view may be observed (created with BioRender.com, accessed on 10 April 2024). Ao VTI—aortic velocity–time integral; Aod—aorta diameter; D1—left ventricle short-axis diameter parallel; D2—left ventricle short-axis diameter perpendicular; HR—heart rate; IVS—intraventricular septum thickness; LA—left atrium; LV—left ventricle; LVET—left ventricle ejection time; LVID—left ventricle internal diameter; LVOT—left ventricular outflow tract; LVPW—left ventricle posterior wall thickness; PA diameter—pulmonary artery; PA VTI—pulmonary artery velocity–time integral; PAAT—pulmonary artery acceleration time; PLAX—parasternal long axis; PSAX—parasternal short axis; RA—right atrium; TAPSE—tricuspid annular plane systolic excursion.</p>
Full article ">Figure 2
<p>Aorta diameter (Aod, mm) measured in 26-week-old female Wistar rats using B-mode at diastole, obtained in PLAX view. The image was obtained using a real-time scanner (Logic P6<sup>®</sup>; General Electric Healthcare, Milwaukee, WI, USA) with a 4–10 MHz linear probe. Measurements were taken using MicroDicom 2023.1 viewer and software.</p>
Full article ">Figure 3
<p>Interventricular septum thickness (IVS, mm), left ventricle internal dimension (LVID), and left ventricle posterior wall thickness (LVPW) measured in 26-week-old female Wistar rats using B-mode in diastole (<b>A</b>) and systole (<b>B</b>), obtained in PLAX view. The images were obtained using a real-time scanner (Logic P6<sup>®</sup>; General Electric Healthcare, Milwaukee, WI, USA) with a 4–10 MHz linear probe. Measurements were taken using MicroDicom 2023.1 viewer and software.</p>
Full article ">Figure 4
<p>Heart rate (HR, bpm) measured in 26-week-old female Wistar rats using M-mode, obtained in PSAX view. The arrows represent the number of systoles captured in 3 seconds. The image was obtained using a real-time scanner (Logic P6<sup>®</sup>; General Electric Healthcare, Milwaukee, WI, USA) with a 4–10 MHz linear probe. Measurements were taken using MicroDicom 2023.1 viewer and software.</p>
Full article ">Figure 5
<p>Left ventricle short-axis diameter parallel (D1) and perpendicular (D2) to the septum measured in 26-week-old female Wistar rats using B-mode at diastole, obtained in PSAX view. The image was obtained using a real-time scanner (Logic P6<sup>®</sup>; General Electric Healthcare, Milwaukee, WI, USA) with a 4–10 MHz linear probe. Measurements were taken using MicroDicom 2023.1 viewer and software.</p>
Full article ">Figure 6
<p>Pulmonary artery acceleration time (PAAT, cm/s) measured in 26-week-old female Wistar rats using Doppler mode, obtained in PSAX view. The image was obtained using a real-time scanner (Logic P6<sup>®</sup>; General Electric Healthcare, Milwaukee, WI, USA) with a 4–10 MHz linear probe. Measurements were taken using MicroDicom 2023.1 viewer and software.</p>
Full article ">Figure 7
<p>Pulmonary artery diameter (mm) measured in 26-week-old female Wistar rats using Doppler mode, obtained in PSAX view. The image was obtained using a real-time scanner (Logic P6<sup>®</sup>; General Electric Healthcare, Milwaukee, WI, USA) with a 4–10 MHz linear probe. Measurements were taken using MicroDicom 2023.1 viewer and software.</p>
Full article ">Figure 8
<p>Tricuspid annular plane systolic excursion (TAPSE, cm) measured in 26-week-old female Wistar rats using M-mode at systole, obtained in apical 4-chamber view. The image was obtained using a real-time scanner (Logic P6<sup>®</sup>; General Electric Healthcare, Milwaukee, WI, USA) with a 4–10 MHz linear probe. Measurements were taken using MicroDicom 2023.1 viewer and software.</p>
Full article ">Figure 9
<p>Peak early diastolic transmitral flow (E, cm/s) and peak late diastolic transmitral flow (A, cm/s) measured in 26-week-old female Wistar rats using Doppler mode at early phase diastole, obtained in apical 4-chamber view. The image was obtained using a real-time scanner (Logic P6<sup>®</sup>; General Electric Healthcare, Milwaukee, WI, USA) with a 4–10 MHz linear probe. Measurements were taken using MicroDicom 2023.1 viewer and software.</p>
Full article ">Figure 10
<p>Right atrium (RA, mm<sup>2</sup>) and left atrium (LA, mm<sup>2</sup>) area measured in 26-week-old female Wistar rats using B-mode, obtained in apical 4-chamber view. The image was obtained using a real-time scanner (Logic P6<sup>®</sup>; General Electric Healthcare, Milwaukee, WI, USA) with a 4–10 MHz linear probe. Measurements were taken using MicroDicom 2023.1 viewer and software.</p>
Full article ">Figure 11
<p>Left ventricle ejection time (LVET, s) duplicate measured in 26-week-old female Wistar rats using Doppler mode, obtained in 5-chamber view. The image was obtained using a real-time scanner (Logic P6<sup>®</sup>; General Electric Healthcare, Milwaukee, WI, USA) with a 4–10 MHz linear probe. Measurements were taken using MicroDicom 2023.1 viewer and software.</p>
Full article ">
24 pages, 2346 KiB  
Article
Multi-Omics Profiles of Small Intestine Organoids in Reaction to Breast Milk and Different Infant Formula Preparations
by Xianli Wang, Shangzhi Yang, Chengdong Zheng, Chenxuan Huang, Haiyang Yao, Zimo Guo, Yilun Wu, Zening Wang, Zhenyang Wu, Ruihong Ge, Wei Cheng, Yuanyuan Yan, Shilong Jiang, Jianguo Sun, Xiaoguang Li, Qinggang Xie and Hui Wang
Nutrients 2024, 16(17), 2951; https://doi.org/10.3390/nu16172951 - 2 Sep 2024
Viewed by 1277
Abstract
Ensuring optimal infant nutrition is crucial for the health and development of children. Many infants aged 0–6 months are fed with infant formula rather than breast milk. Research on cancer cell lines and animal models is limited to examining the nutrition effects of [...] Read more.
Ensuring optimal infant nutrition is crucial for the health and development of children. Many infants aged 0–6 months are fed with infant formula rather than breast milk. Research on cancer cell lines and animal models is limited to examining the nutrition effects of formula and breast milk, as it does not comprehensively consider absorption, metabolism, and the health and social determinants of the infant and its physiology. Our study utilized small intestine organoids induced from human embryo stem cell (ESC) to compare the nutritional effects of breast milk from five donors during their postpartum lactation period of 1–6 months and three types of Stage 1 infant formulae from regular retail stores. Using transcriptomics and untargeted metabolomics approaches, we focused on the differences such as cell growth and development, cell junctions, and extracellular matrix. We also analyzed the roles of pathways including AMPK, Hippo, and Wnt, and identified key genes such as ALPI, SMAD3, TJP1, and WWTR1 for small intestine development. Through observational and in-vitro analysis, our study demonstrates ESC-derived organoids might be a promising model for exploring nutritional effects and underlying mechanisms. Full article
(This article belongs to the Topic Advances in Animal-Derived Non-Cow Milk and Milk Products)
Show Figures

Figure 1

Figure 1
<p>Transcriptome profiles of intestine organoids feeding by different infant formulae and breast milk. (<b>a</b>) PCA of samples in groups BM, PMF1, PMF2, PMF3, and control. PC1 and PC2 Scores of different samples are visualized, and the variance contributed by its corresponding component is presented. (<b>b</b>) GSVA analysis of each sample for GO terms associated with nutrition absorption in small intestine. (<b>c</b>) Venn graph of different groups’ DEG. (<b>d</b>,<b>e</b>) Gene over-representation analysis of GO of (<b>d</b>) unique DEG of infant formulae group and breast milk group presented in functionally grouped network with terms as nodes linked based on their kappa score level (≥0.3) using a Cytoscape plug-in clueGO, and (<b>e</b>) shared DEG of all infant formulae and breast milk presented in dendrogram using methods adopted by GeneTonic. (<b>f</b>) Pathway enrichment analysis of KEGG for shared DEG of all infant formulae and breastmilk. Top 12 enriched significant pathways (<span class="html-italic">p</span> value &lt; 0.05) ordered by count were presented.</p>
Full article ">Figure 2
<p>Metabolite profiles of breast milk and different infant formulae. (<b>a</b>) Inter-group PLSDA (Partial Least Square Discriminant Analysis). Variation contribution of each component was presented. (<b>b</b>) A hierarchical clustered heatmap of different metabolites identified by ANOVA analysis. (<b>c</b>) Venn plot of enriched pathways of differential metabolites of BM, PMF1, PMF2, and PMF3. (<b>d</b>) KEGG pathways enriched from differential metabolites of BM, PMF1, PMF2, and PMF3. Pathways satisfying <span class="html-italic">p</span> value &lt; 0.1 were presented. (<b>e</b>) KEGG pathways enriched from differential metabolites of different breast milk groups. Pathways satisfying <span class="html-italic">p</span> value &lt; 0.1 were presented.</p>
Full article ">Figure 3
<p>Pro-development effects of breast milk and different infant formulae on intestine organoids. (<b>a</b>) Radar plot of GO-enriched pathways’ z-score of breast milk group and PMF group. Methods are from GeneTonic, an R package for RNA-seq data. Z-score implies the intensity and direction of pathway enrichment. PMF means powder milk (infant formulae), gathering PMF1, PMF2, and PMF3 as one group. (<b>b</b>) GSEA results of GO: Canonical Wnt signaling pathway from shared DEG of BM, PMF1, PMF2, and PMF3. NES means normalized enrichment score. (<b>c</b>) Heatmap of core enrichment genes of GSEA: canonical Wnt signaling pathway. (<b>d</b>) Heatmap of GSVA score of selected pathways associated with growth and development of intestine for each sample. (<b>e</b>) Cilium assembly and epithelial tube formation’ highly correlated genes (GSVA score-gene, spearman correlation &gt; 0.8) and the metabolites highly correlated to them (gene-metabolite, spearman correlation &gt; 0.8). (<b>f</b>) mRNA expression level of ‘key genes’ measured by RT-qPCR technique (2<sup>−ΔΔCt</sup> method). All data are presented as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001; ns, no significance, <span class="html-italic">p</span> ≥ 0.05.</p>
Full article ">Figure 4
<p>Breast milk and different infant formulae effects on cell junction assembly and regulation. (<b>a</b>) NES value of GSEA enrichment for breast milk and infant formulae. Here, treat infant formulae as one group: PMF. (<b>b</b>) Inter-PMF comparison of cell junction-related GO biological processes enriched by GSEA. NES of each GO term for each infant formula group was visualized. (<b>c</b>) mRNA level of claudin-1 (CLDN1) and ZO-1 (TJP1) quantified by RT-qPCR (2<sup>−ΔΔCt</sup> method). (<b>d</b>) Hierarchical clustered heatmap of expression of core enrichment genes in tight junction pathway. Genes were clustered in three modules, representing certain infant formula groups’ relatively higher expressed genes. (<b>e</b>) Protein–protein interactions, respectively, from each clustered module’s genes. Left-Up: from module representing PMF1. Right-Up: from module representing PMF3. Middle-Down: from module representing PMF2. All data are presented as mean ± SEM. ** <span class="html-italic">p</span> &lt; 0.01; ns, no significance, <span class="html-italic">p</span> ≥ 0.05.</p>
Full article ">Figure 5
<p>Profiles of extracellular events of breast milk and infant formulae. (<b>a</b>) GSEA result for GO (CC, cell compartment): Extracellular matrix obtained from identified shared DEG of breast milk and different infant formulae. (<b>b</b>) Heatmap of expression of core enrichment genes in Extracellular Matrix (ECM) generated from GSEA. (<b>c</b>) “Hub genes” and their highly correlated metabolites identified from network of ECM-related DEGs of BM. (<b>d</b>) “Hub genes” and their highly correlated metabolites identified from network of ECM-related DEGs of PMF. (<b>e</b>) Major GO terms enriched from ECM-related DEG of BM in the form of functionally grouped networks. (<b>f</b>) Major GO terms enriched from ECM-related DEG of PMF in the form of functionally grouped networks, similar terms were fused by clueGO.</p>
Full article ">
8 pages, 865 KiB  
Commentary
GDF15 Targeting for Treatment of Hyperemesis Gravidarum
by Jamie Thygerson, Dallin Oyler, Jackson Thomas, Brandon Muse, Benjamin D. Brooks and Jessica E. Pullan
Medicines 2024, 11(7), 17; https://doi.org/10.3390/medicines11070017 - 30 Aug 2024
Viewed by 270
Abstract
Nausea and vomiting during pregnancy (NVP), particularly its severe form, Hyperemesis gravidarum (HG), affects up to 70% of pregnancies and significantly impacts the quality of life for those with the condition as well as generates a great economic burden, with annual costs exceeding [...] Read more.
Nausea and vomiting during pregnancy (NVP), particularly its severe form, Hyperemesis gravidarum (HG), affects up to 70% of pregnancies and significantly impacts the quality of life for those with the condition as well as generates a great economic burden, with annual costs exceeding $1.7 billion in the United States. Despite the available treatments targeting neurotransmitters like serotonin and dopamine, many patients experience inadequate relief and suffer from severe side effects, including headaches and dizziness. Recent research has underscored the role of GDF15, a protein mainly produced by the placenta and linked to NVP symptoms. This protein, part of the TGF-β superfamily, has been implicated in appetite and weight regulation and is altered in those with HG due to specific genetic mutations. Addressing the challenges of delivering effective treatments, current innovations focus on targeting GDF15 to reduce symptoms while ensuring fetal safety. Promising therapeutic strategies include non-IgG immunotherapies, small peptide and molecule antagonists, and novel administration methods such as transdermal patches. These approaches aim to optimize dosage and reduce adverse effects. The effective development and testing of these treatments necessitate advanced animal models that closely resemble human pregnancy physiology, highlighting the need for further research and funding. This ongoing research holds significant potential to improve the clinical outcomes for HG patients and decrease the economic impact on healthcare systems, urging a dedicated response from the scientific and medical communities to advance these promising treatments. Full article
Show Figures

Figure 1

Figure 1
<p>The mechanism of action of GDF15 and GFRAL in the brain. GDF15 from the placenta enters the area postrema to bind GFRAL in the area postrema, activating RET and downstream pathways (PI3K/AKT, PLC/PKC, MEK/ERK). This signaling impacts adipose tissue (increasing fatty acid oxidation and weight loss), the liver (reducing glucose and cholesterol), and the stomach (decreasing food intake and gastric emptying, inducing weight loss and taste aversion). Green arrows indicate increase, red arrows indicate decrease.</p>
Full article ">Figure 2
<p>Illustrates the inhibition of GDF15–GFRAL binding and subsequent RET activation. The top section highlights the area postrema in the brain, where GDF15 binds to GFRAL, leading to RET activation (shown in detail in the top box). The bottom box details four therapeutic strategies: (<b>A</b>) an antibody that binds to GFRAL, blocking GDF15 interaction and preventing RET activation; (<b>B</b>) a small peptide antagonist that binds to GFRAL after GDF15 binds, preventing the attachment of RET; (<b>C</b>) a small molecule inhibitor that binds directly to GDF15, preventing its interaction with GFRAL, or binding to and inhibiting GFRAL and RET externally; and (<b>D</b>) a small molecule delivered via a nanoparticle to enhance targeting and reduce side effects. Each strategy aims to disrupt the GDF15–GFRAL interaction in a targeted approach.</p>
Full article ">
12 pages, 4159 KiB  
Article
Longitudinal Imaging of Injured Spinal Cord Myelin and White Matter with 3D Ultrashort Echo Time Magnetization Transfer (UTE-MT) and Diffusion MRI
by Qingbo Tang, Yajun Ma, Qun Cheng, Yuanshan Wu, Junyuan Chen, Jiang Du, Pengzhe Lu and Eric Y. Chang
J. Imaging 2024, 10(9), 213; https://doi.org/10.3390/jimaging10090213 - 30 Aug 2024
Viewed by 400
Abstract
Quantitative MRI techniques could be helpful to noninvasively and longitudinally monitor dynamic changes in spinal cord white matter following injury, but imaging and postprocessing techniques in small animals remain lacking. Unilateral C5 hemisection lesions were created in a rat model, and ultrashort echo [...] Read more.
Quantitative MRI techniques could be helpful to noninvasively and longitudinally monitor dynamic changes in spinal cord white matter following injury, but imaging and postprocessing techniques in small animals remain lacking. Unilateral C5 hemisection lesions were created in a rat model, and ultrashort echo time magnetization transfer (UTE-MT) and diffusion-weighted sequences were used for imaging following injury. Magnetization transfer ratio (MTR) measurements and preferential diffusion along the longitudinal axis of the spinal cord were calculated as fractional anisotropy or an apparent diffusion coefficient ratio over transverse directions. The area of myelinated white matter was obtained by thresholding the spinal cord using mean MTR or diffusion ratio values from the contralesional side of the spinal cord. A decrease in white matter areas was observed on the ipsilesional side caudal to the lesions, which is consistent with known myelin and axonal changes following spinal cord injury. The myelinated white matter area obtained through the UTE-MT technique and the white matter area obtained through diffusion imaging techniques showed better performance to distinguish evolution after injury (AUCs > 0.94, p < 0.001) than the mean MTR (AUC = 0.74, p = 0.01) or ADC ratio (AUC = 0.68, p = 0.05) values themselves. Immunostaining for myelin basic protein (MBP) and neurofilament protein NF200 (NF200) showed atrophy and axonal degeneration, confirming the MRI results. These compositional and microstructural MRI techniques may be used to detect demyelination or remyelination in the spinal cord after spinal cord injury. Full article
(This article belongs to the Section Medical Imaging)
Show Figures

Figure 1

Figure 1
<p>3D UTE-MT pulse sequence diagram. A Fermi-shaped pulse (duration =8 msec and bandwidth = 160 Hz) was used to generate MT contrast with three different flip angles of 1500°, 800°, and 0°, and a frequency offset of 1500 Hz.</p>
Full article ">Figure 2
<p>Longitudinal MTR mapping results of a representative rat at a location 5 mm caudal to unilateral C5 hemisectioning (columns 1 and 2) and the corresponding results from a control rat (columns 3 and 4). Decreases in MTR on the ipsilesional side are seen on the whole cord MTR maps of the injured rat beginning at 8 weeks (arrows). Myelinated white matter maps, which were obtained by thresholding using the mean MTR of the contralesional side on the same slice, show a markedly decreased total area on the injured side (arrowheads).</p>
Full article ">Figure 3
<p>Time course of changes in MTR (<b>A</b>) and the myelinated white matter areas (<b>B</b>) of the contralesional side (contra), ipsilesional side (ipsi), and control rats at a location 5 mm caudal to a C5 hemisection. * <span class="html-italic">p</span> &lt; 0.01, # <span class="html-italic">p</span> &lt; 0.001 when the ipsilesional side was compared with the contralesional side using a two-way ANOVA followed by Tukey’s multiple comparisons test.</p>
Full article ">Figure 4
<p>Longitudinal diffusion maps of a representative rat at a location 5 mm caudal to unilateral C5 hemisectioning in columns 1 to 5. Diffusivities in the radial (ADC_X and ADC_Y), axial (ADC_Z), and axial-over-radial diffusion directions (ADC_R) are shown as white matter maps, which were obtained by thresholding using the mean ADC_R value of the contralesional side on the same slice. The white matter maps of a control rat similarly obtained by thresholding are shown in the last column.</p>
Full article ">Figure 5
<p>Time course of changes in fractional anisotropy (<b>A</b>), ADC_R (<b>B</b>), and the white matter area (<b>C</b>) of the contralesional side (contra), ipsilesional side (ipsi), and control rats at a location 5 mm caudal to a C5 hemisection. * <span class="html-italic">p</span> &lt; 0.05, # <span class="html-italic">p</span> &lt; 0.01, ## <span class="html-italic">p</span> &lt; 0.0001 when the ipsilesional side was compared with the contralesional side with a two-way ANOVA followed by Tukey’s multiple comparisons test.</p>
Full article ">Figure 6
<p>The receiver operator characteristic curve (ROC) analyses comparing MTR and the myelinated white matter area derived from thresholding of MTR (<b>A</b>), and comparing ADC_R and the white matter areas derived from thresholding of ADC_R (<b>B</b>). Data were from the contralesional and ipsilesional sides of five rats at week 2, 8, 14, and 20 post-injury.</p>
Full article ">Figure 7
<p>Immunofluorescence images after immunostaining for myelin basic protein (MBP, green, top) and NF200 (red, bottom) in a spinal cord section about 5 mm caudal to a unilateral C5 hemisection on the right side 20 weeks post-injury, showing atrophy and decreased staining intensity on the ipsilesional side.</p>
Full article ">
12 pages, 3025 KiB  
Article
The Skin Histopathology of Pro- and Parabiotics in a Mouse Model of Atopic Dermatitis
by Hun Hwan Kim, Se Hyo Jeong, Min Yeong Park, Pritam Bhagwan Bhosale, Abuyaseer Abusaliya, Jeong Doo Heo, Hyun Wook Kim, Je Kyung Seong, Tae Yang Kim, Jeong Woo Park, Byeong Soo Kim and Gon Sup Kim
Nutrients 2024, 16(17), 2903; https://doi.org/10.3390/nu16172903 - 30 Aug 2024
Viewed by 560
Abstract
As it has been revealed that the activation of human immune cells through the activity of intestinal microorganisms such as pro- and prebiotics plays a vital role, controlling the proliferation of beneficial bacteria and suppressing harmful bacteria in the intestine has become essential. [...] Read more.
As it has been revealed that the activation of human immune cells through the activity of intestinal microorganisms such as pro- and prebiotics plays a vital role, controlling the proliferation of beneficial bacteria and suppressing harmful bacteria in the intestine has become essential. The importance of probiotics, especially for skin health and the immune system, has led to the emergence of products in various forms, including probiotics, prebiotics, and parabiotics. In particular, atopic dermatitis (AD) produces hypersensitive immunosuppressive substances by promoting the differentiation and activity of immune regulatory T cells. As a result, it has been in the Th1 and Th2 immune balance through a mechanism that suppresses skin inflammation or allergic immune responses caused by bacteria. Furthermore, an immune mechanism has recently emerged that simultaneously controls the expression of IL-17 produced by Th17. Therefore, the anti-atopic effect was investigated by administering doses of anti-atopic candidate substances (Lactobacilus sakei CVL-001, Lactobacilus casei MCL, and Lactobacilus sakei CVL-001 Lactobacilus casei MCL mixed at a ratio of 4:3) in an atopy model using 2,4-dinitrochlorobenzene and observing symptom changes for 2 weeks to confirm the effect of pro-, para-, and mixed biotics on AD. First, the body weight and feed intake of the experimental animals were investigated, and total IgG and IgM were confirmed through blood biochemical tests. Afterward, histopathological staining was performed using H&E staining, Toluidine blue staining, Filaggrin staining, and CD8 antibody staining. In the treatment group, the hyperproliferation of the epidermal layer, the inflammatory cell infiltration of the dermal layer, the expression of CD8, the expression of filaggrin, and the secretion of mast cells were confirmed to be significantly reduced. Lastly, small intestine villi were observed through a scanning microscope, and scoring evaluation was performed through skin damage. Through these results, it was confirmed that AD was reduced when treated with pro-, para-, and mixed biotics containing probiotics and parabiotics. Full article
(This article belongs to the Special Issue The Role of Probiotics and Prebiotics in Immunomodulation)
Show Figures

Figure 1

Figure 1
<p>Histologic features of DNCB-induced AD-like skin damage were evaluated using H&amp;E staining. (<b>A</b>) H&amp;E staining of experimental animal skin for anti-atopic effects of pro-, para-, and mixed biotics (H&amp;E × 200). (<b>B</b>) Histopathologic score in AD-like skin lesions. H&amp;E staining showed that pro-, para-, and mixed biotics all alleviated DNCB-induced inflammation in the epidermal layer, with a histologically significant reduction in the T3 group. The red line in the figure is the relative width of the tissue stain. The data represent the mean ± SD of three independent experiments. (## <span class="html-italic">p</span> &lt; 0.01 vs. VC, * <span class="html-italic">p</span> &lt; 0.05 vs. NC, ** <span class="html-italic">p</span> &lt; 0.01 vs. NC).</p>
Full article ">Figure 2
<p>Histologic features of CD8 antibody staining and Filaggrin staining of AD-like skin lesions with DNCB. (<b>A</b>) CD8 antibody staining of experimental animal skin for anti-atopic effects of pro-, para-, and mixed biotics. Increased expression of CD8 in the epidermal and dermal layers was observed in the DNCB-induced NC, but relatively decreased CD8 expression was observed in the T1, T2, and T3 groups. Similarly, (<b>B</b>) Filaggrin staining in AD showed an increase in the thickness of the epithelial layer in the DNCB-induced NC, but a decrease in T1, T2, and T3. The data represent the mean ± SD of three independent experiments. (# <span class="html-italic">p</span> &lt; 0.05 vs. VC, ## <span class="html-italic">p</span> &lt; 0.01 vs. VC, * <span class="html-italic">p</span> &lt; 0.05 vs. NC, ** <span class="html-italic">p</span> &lt; 0.01 vs. NC, *** <span class="html-italic">p</span> &lt; 0.001 vs. NC).</p>
Full article ">Figure 3
<p>Toluidine blue staining histologic features of AD-like skin lesions with DNCB. (<b>A</b>) Toluidine blue staining of experimental animal skin for anti-atopic effects of pro-, para-, and mixed biotics. (<b>B</b>) Mast cell count in AD-like skin lesions. Mast cells increased dramatically in the DNCB-induced group, but decreased in the T1, T2, and T3 groups, with a significant decrease in the T3 group. The red arrow is the estimated number of mast cells. Data represent the mean ± SD of three independent experiments. (## <span class="html-italic">p</span> &lt; 0.01 vs. VC, * <span class="html-italic">p</span> &lt; 0.05 vs. NC, ** <span class="html-italic">p</span> &lt; 0.01 vs. NC).</p>
Full article ">Figure 4
<p>(<b>A</b>) Visual assessment and average atopic score for each symptom between days 1–15 of atopy-induced skin damage with DNCB and scratching behavior scoring results. (<b>B</b>) Enlarged the condition of atopic dermatitis in each group. (<b>C</b>) Each item is scored as no symptoms (0), mild (1), moderate (2), or severe (3). Means with different superscripts in the same row are significantly different at <span class="html-italic">p</span> &lt; 0.05 via Duncan’s multiple range tests. The data represent the mean ± SD of three independent experiments. (# <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01 vs. VC).</p>
Full article ">
28 pages, 388 KiB  
Review
Feeding Values of Indigenous Browse Species and Forage Legumes for the Feeding of Ruminants in Ethiopia: A Meta-Analysis
by Sisay Belete, Adugna Tolera, Simret Betsha and Uta Dickhöfer
Agriculture 2024, 14(9), 1475; https://doi.org/10.3390/agriculture14091475 - 29 Aug 2024
Viewed by 409
Abstract
The foliage of browse species and forage legumes has good nutritional value and can be utilized as a protein source in ruminant diets. However, its efficient utilization requires the establishment of a comprehensive database of feeding values. Two databases, i.e., forage nutritive value [...] Read more.
The foliage of browse species and forage legumes has good nutritional value and can be utilized as a protein source in ruminant diets. However, its efficient utilization requires the establishment of a comprehensive database of feeding values. Two databases, i.e., forage nutritive value (92 studies) and in vivo animal performance (62 feeding experiments), were built to assess the feeding value of the foliage of browse species and cultivated forage legumes in Ethiopia. The forage nutritive value data (chemical composition and in vitro digestibility) were summarized as descriptive statistics. The analysis of in vivo data was conducted using a mixed model procedure with fixed (forage supplement) and random (studies) factors. Forage categories had crude protein (CP) ranging from 17.6 ± 5.2% (indigenous browse species) to 22.4 ± 4.5% (multipurpose fodder tree/shrub species), respectively. Variations were observed in CP values between the vegetative and blooming stage harvesting of herbaceous forages (22.7 ± 4.1% versus 19.8 ± 3.5%). The leaves contained more CP than the twigs in multipurpose fodder tree/shrubs (22.8 ± 3.2% versus 18.8 ± 0.6%) and the pods in indigenous browse species (18.0 ± 5.0% versus 15.3 ± 2.3%). However, the greatest mean in vitro organic matter digestibility (IVOMD) of 70.1 ± 10.8% was observed in the foliage of indigenous browse species. The variation in IVOMD was small among the forage categories (61.2 ± 11.2%–63.5 ± 10.8%). Twigs of the multipurpose fodder tree/shrub species had the lowest IVOMD of 53.0 ± 6.9%. Herbaceous forage legumes tended to have higher NDF and ADF values than the other forage categories. In terms of nutrient concentration and digestibility, large variations were observed within the same forage categories and species. The supplementation of forage, on average at 277.5 ± 101.4 g/day (±SD), to a low-quality basal diet resulted in a significant (p < 0.05) improvement in the apparent digestibility of DM, CP, and NDF as well as the daily intake of DM, CP, and metabolizable energy (ME). The application of sole forage supplementation was determined to have comparable effects on DM intake (p = 0.2347) with dietary supplements based on concentrate feedstuffs. However, CP intake (p = 0.0733) tended to be lower for forage over the concentrate treatment. The averaged daily gain (ADG) of the animals was significantly increased (p < 0.05) by 71.2% due to the forage supplement compared to unsupplemented treatment (11.6 ± 5.47 g/d (±SE) vs. 40.3 ± 4.99 g/d (±SE)). Overall, the nutrient utilization and production performance of animals fed with low-quality basal diets could be improved when an appropriate amount of forage is included as supplement. The large variation recorded in the nutritional composition of browse species and forage legumes could provide an opportunity to screen for species and varieties with superior nutritional quality. Full article
31 pages, 2559 KiB  
Review
Origami of KR-12 Designed Antimicrobial Peptides and Their Potential Applications
by Jayaram Lakshmaiah Narayana, Abraham Fikru Mechesso, Imran Ibni Gani Rather, D. Zarena, Jinghui Luo, Jingwei Xie and Guangshun Wang
Antibiotics 2024, 13(9), 816; https://doi.org/10.3390/antibiotics13090816 - 28 Aug 2024
Viewed by 1041
Abstract
This review describes the discovery, structure, activity, engineered constructs, and applications of KR-12, the smallest antibacterial peptide of human cathelicidin LL-37, the production of which can be induced under sunlight or by vitamin D. It is a moonlighting peptide that shows both antimicrobial [...] Read more.
This review describes the discovery, structure, activity, engineered constructs, and applications of KR-12, the smallest antibacterial peptide of human cathelicidin LL-37, the production of which can be induced under sunlight or by vitamin D. It is a moonlighting peptide that shows both antimicrobial and immune-regulatory effects. Compared to LL-37, KR-12 is extremely appealing due to its small size, lack of toxicity, and narrow-spectrum antimicrobial activity. Consequently, various KR-12 peptides have been engineered to tune peptide activity and stability via amino acid substitution, end capping, hybridization, conjugation, sidechain stapling, and backbone macrocyclization. We also mention recently discovered peptides KR-8 and RIK-10 that are shorter than KR-12. Nano-formulation provides an avenue to targeted delivery, controlled release, and increased bioavailability. In addition, KR-12 has been covalently immobilized on biomaterials/medical implants to prevent biofilm formation. These constructs with enhanced potency and stability are demonstrated to eradicate drug-resistant pathogens, disrupt preformed biofilms, neutralize endotoxins, and regulate host immune responses. Also highlighted are the safety and efficacy of these peptides in various topical and systemic animal models. Finaly, we summarize the achievements and discuss future developments of KR-12 peptides as cosmetic preservatives, novel antibiotics, anti-inflammatory peptides, and microbiota-restoring agents. Full article
(This article belongs to the Special Issue Insights into Natural Antimicrobial Peptides)
Show Figures

Figure 1

Figure 1
<p>Properties and functions of human cathelicidin LL-37 discovered in different cells. Depicted in the center is the 3D structure of membrane-bound LL-37 (PDB ID: 2K6O) determined by 3D triple-resonance heteronuclear multidimensional nuclear magnetic resonance (NMR) spectroscopy. When targeting bacterial membranes, the C-terminal tail of LL-37 is not folded and remains highly flexible as confirmed by heteronuclear <sup>15</sup>N backbone dynamics on the ps-ns time scale [<a href="#B32-antibiotics-13-00816" class="html-bibr">32</a>]. The C-terminal tail is disordered in complex with SDS, D8PG, and LPS (abbreviations in the text). Direct interactions of LL-37 with anionic bacterial phosphatidylglycerols (PGs) and LPS as demonstrated by NMR provide basis for antimicrobial and anti-inflammatory effects. NET: neutrophil extracellular traps; PBMC: peripheral blood mononuclear cells; DCs: dendritic cells; MSC: mesenchymal stem cell.</p>
Full article ">Figure 2
<p>Therapeutic strategies based on human cathelicidin LL-37. (<b>A</b>) Humans can use sunlight or vitamin D and its analog to switch on the expression of LL-37 to boost innate defense against infection. Likewise, recombinant DNA technology can be used to express LL-37 to achieve the same production. (<b>B</b>) Human LL-37 can function synergistically with other human AMPs such as defensin or lysozyme to better control pathogens. Similarly, human LL-37 can work synergistically with bacteriocins from commensal bacteria to better control invading pathogens. Using the same strategy, AMPs can be used with existing antibiotics to overcome resistance. (<b>C</b>) LL-37 can be engineered into novel antimicrobial agents based on different fragments (IG-24, GF-17, and KR-12) discovered from (1) peptide library, (2) structure-based design, (3) combined (1) and (2), and (4) feature-based mimicking (reviewed in ref. [<a href="#B26-antibiotics-13-00816" class="html-bibr">26</a>]). This review focuses on a variety of the engineered constructs based on KR-12, the smallest antibacterial fragment of LL-37.</p>
Full article ">Figure 3
<p>The discovery path of KR-12 through structural studies. (<b>A</b>) Amino acid sequences and nomenclature of LL-37 and its fragments. The original peptide names for these fragments are given on the right of the peptide sequences, while the shortened names are provided on the left. While LL-37 has a carboxylic acid at the C-terminus, the C-termini of shorter sequences, including FK-16/GF-17, FK-13, and KR-12, are all amidated to increase the net charge by +1. (<b>B</b>) Backbone structures of LL-37, IG-25, GF-17, FK-13, and KR-12 determined by 2D and 3D NMR spectroscopy [<a href="#B32-antibiotics-13-00816" class="html-bibr">32</a>,<a href="#B39-antibiotics-13-00816" class="html-bibr">39</a>,<a href="#B60-antibiotics-13-00816" class="html-bibr">60</a>]. Except for LL-37 and IG-25, GF-17, FK-13, and KR-12 are C-terminally amidated. (<b>C</b>,<b>D</b>) Horizontal and vertical views of the NMR structure of KR-12 in complex with anionic D8PG [<a href="#B32-antibiotics-13-00816" class="html-bibr">32</a>].</p>
Full article ">Figure 4
<p>Helical wheel plots for LL-37 (<b>A</b>), KR-12 (<b>B</b>) and its selective derivatives (<b>C</b>–<b>L</b>). The helical wheel was generated using the NetWheel program (<a href="http://lbqp.unb.br/NetWheels/" target="_blank">http://lbqp.unb.br/NetWheels/</a>, accessed on 31 July 2024). In this program, amino acids are classified into four groups: (1) polar/basic (red square): RHK, (2) polar/acidic (blue triangle): DE, (3) polar/uncharged (green diamond): STNQC, and (4) nonpolar (yellow circle): AGVILMFYWP. Although included in the plot, it is evident that the HIV TAT sequence is not amphipathic (<b>J</b>). However, the amphipathic helical structure can still be seen in the presence of additional sequence from the Trp cage (<b>K</b>). Finally, some symmetry can be seen in the helical wheel plot of cyclic KR-12 dimer (<b>L</b>).</p>
Full article ">Figure 5
<p>Various design strategies that transform human LL-37-derived KR-12 to new constructs: (1) amino acid changes, terminal capping, peptide hybridization, and peptide conjugates (<b>top</b>), (2) sidechain stapling and backbone cyclization (<b>right</b>), (3) surface immobilization (<b>left</b>), and (4) peptide formulation (<b>bottom</b>). See the text for further details. These strategies can be applied to other linear antimicrobial peptides as well.</p>
Full article ">Figure 6
<p>2D array of the lengths of the KR-12 peptide with 4 to 12 amino acids (aa4 to aa12) and fatty acids (c6 to C14) for antibacterial activity (<b>A</b>) and hemolytic toxicity (<b>B</b>) uncovered a zone for designing selective lipopeptides. On the left, the closer the curves to the green plane, the more potent the peptides are. In contrast, the farther away from the red plane on the right, the less hemolytic the peptides are.</p>
Full article ">Figure 7
<p>Like LL-37, KR-12 peptides also possess numerous desired properties such as antimicrobial, antibiofilm, and LPS neutralization. KR-12, as well as LL-37, has been covalently immobilized onto titanium (Ti) implants [<a href="#B123-antibiotics-13-00816" class="html-bibr">123</a>,<a href="#B124-antibiotics-13-00816" class="html-bibr">124</a>].</p>
Full article ">Figure 8
<p>In vivo safety and efficacy of KR-12 constructs. (<b>A</b>) Systemic toxicity of nanobiotics Myr-KR-12N and Myr-KR-12C via intravenous administration in mice. (<b>B</b>) Ototoxicity of a KR-12-a2 by applying the solution topically into the middle ears of guinea pigs. (<b>C</b>) Identification of the non-toxic dose of C10-KR8d via the intraperitoneal route in mice. (<b>D</b>) Efficacy of Ti-C10-KR8d implant on catheter-associated MRSA biofilm in mice. (<b>E</b>) LPS neutralization and bone restoration efficacy of KR-12-a2 in mice. (<b>F</b>) Cryogel-HA/TA/KR12 topical application in a mouse wound model. (<b>G</b>) PEEK-PDA-KR-12 coating on implants shows both antibacterial and osteointegration potential in mice. (<b>H</b>) KR-12 has anti-colitis ability against chemical induced colitis in mice. (<b>I</b>) Myr-KR-12N and Myr-KR-12C protection from LPS sepsis in mice. (<b>J</b>) C10-KR8d showcased anti-MRSA efficacy in a neutropenic murine infection model.</p>
Full article ">
15 pages, 1007 KiB  
Article
Effect of Selol on Tumor Morphology and Biochemical Parameters Associated with Oxidative Stress in a Prostate Tumor-Bearing Mice Model
by Małgorzata Sochacka, Grażyna Hoser, Małgorzata Remiszewska, Piotr Suchocki, Krzysztof Sikora and Joanna Giebułtowicz
Nutrients 2024, 16(17), 2860; https://doi.org/10.3390/nu16172860 - 27 Aug 2024
Viewed by 526
Abstract
Prostate cancer is the leading cause of cancer death in men. Some studies suggest that selenium Se (+4) may help prevent prostate cancer. Certain forms of Se (+4), such as Selol, have shown anticancer activity with demonstrated pro-oxidative effects, which can lead to [...] Read more.
Prostate cancer is the leading cause of cancer death in men. Some studies suggest that selenium Se (+4) may help prevent prostate cancer. Certain forms of Se (+4), such as Selol, have shown anticancer activity with demonstrated pro-oxidative effects, which can lead to cellular damage and cell death, making them potential candidates for cancer therapy. Our recent study in healthy mice found that Selol changes the oxidative–antioxidative status in blood and tissue. However, there are no data on the effect of Selol in mice with tumors, considering that the tumor itself influences this balance. This research investigated the impact of Selol on tumor morphology and oxidative–antioxidative status in blood and tumors, which may be crucial for the formulation’s effectiveness. Our study was conducted on healthy and tumor-bearing animal models, which were either administered Selol or not. We determined antioxidant enzyme activities (Se-GPx, GPx, GST, and TrxR) spectrophotometrically in blood and the tumor. Furthermore, we measured plasma prostate-specific antigen (PSA) levels, plasma and tumor malondialdehyde (MDA) concentration as a biomarker of oxidative stress, selenium (Se) concentrations and the tumor ORAC value. Additionally, we assessed the impact of Selol on tumor morphology and the expression of p53, BCL2, and Ki-67. The results indicate that treatment with Selol influences the morphology of tumor cells, indicating a potential role in inducing cell death through necrosis. Long-term supplementation with Selol increased antioxidant enzyme activity in healthy animals and triggered oxidative stress in cancer cells, activating their antioxidant defense mechanisms. This research pathway shows promise in understanding the anticancer effects of Selol. Selol appears to increase the breakdown of cancer cells more effectively in small tumors than in larger ones. In advanced tumors, it may accelerate tumor growth if used as monotherapy. Therefore, further studies are necessary to evaluate its efficacy either in combination therapy or for the prevention of recurrence. Full article
(This article belongs to the Special Issue Effects of Selenium and Other Micronutrient Intake on Human Health)
Show Figures

Figure 1

Figure 1
<p>The mean enzyme activities of selenium-dependent glutathione peroxidase (Se-GPx) and total glutathione peroxidase (GPx) (<b>a</b>), and glutathione S-transferase (GST) and thioredoxin reductase (TrxR) (<b>b</b>); concentration of malondialdehyde (MDA) (<b>c</b>), ORAC value (<b>d</b>), and selenium (Se) concentration (<b>e</b>) in the tumor tissue of mice with xenografted LNCaP prostate cancer supplemented with Selol (Ca + Se) and control group of mice with xenografted LNCaP prostate supplemented with placebo (Ca). Data are shown as means ± SD (<span class="html-italic">n</span> = 6). The <span class="html-italic">p</span>-values indicate differences between control group (Ca) and study group (Ca + Se) and are indicated by * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>Relationship between PSA concentrations at the beginning (PSA1) and end (PSA2) of the experiment in tumor-bearing mice treated with Selol (Ca + Se, <span class="html-italic">n</span> = 6) or placebo (Ca, <span class="html-italic">n</span> = 6). Notably, two mice in the Ca + Se group significantly deviated from the expected trend (indicated by red loops) (<b>a</b>). Relationship between PSA concentration at the end of the experiment (PSA2) and tumor mass in tumor-bearing mice treated with Selol (Ca + Se, <span class="html-italic">n</span> = 6) or placebo (Ca, <span class="html-italic">n</span> = 6). Two mice in the Ca + Se group showed significant deviations from the overall trend (indicated by red loops) (<b>b</b>).</p>
Full article ">Figure 3
<p>Histological photomicrographs of tumor sections stained with (<b>a</b>–<b>d</b>) H&amp;E (hematoxylin and eosin): no features of necrosis (<b>a</b>); features of necrosis (<b>b</b>); lite tumor tissue (<b>c</b>) reduction in tumor foci (<b>d</b>); (<b>e</b>,<b>f</b>) stained with p53 (no gene expression in both cases); (<b>g</b>,<b>h</b>) stained with Ki-67 (features of high proliferation activity in both cases); and (<b>i</b>,<b>j</b>) stained with BCL2 (no gene expression in both cases). Ca + Se—mice with xenografted LNCaP prostate cancer supplemented with Selol; Ca—control group of mice with xenografted LNCaP prostate supplemented with placebo.</p>
Full article ">
16 pages, 14420 KiB  
Article
Characterization and Functional Analysis of the 17-Beta Hydroxysteroid Dehydrogenase 2 (hsd17b2) Gene during Sex Reversal in the Ricefield Eel (Monopterus albus)
by Ruyi Chen, Haoyu Zhu, Xiaoling Zhang, Lingli Li, Jinglin Xu, Zhimin Tan, Jialin Su, Ke Feng, Kaili Chen and Hongyan Xu
Int. J. Mol. Sci. 2024, 25(16), 9063; https://doi.org/10.3390/ijms25169063 - 21 Aug 2024
Viewed by 491
Abstract
In mammals, 17-beta hydroxysteroid dehydrogenase 2 (Hsd17b2) enzyme specifically catalyzes the oxidation of the C17 hydroxyl group and efficiently regulates the activities of estrogens and androgens to prevent diseases induced by hormone disorders. However, the functions of the hsd17b2 gene involved in animal [...] Read more.
In mammals, 17-beta hydroxysteroid dehydrogenase 2 (Hsd17b2) enzyme specifically catalyzes the oxidation of the C17 hydroxyl group and efficiently regulates the activities of estrogens and androgens to prevent diseases induced by hormone disorders. However, the functions of the hsd17b2 gene involved in animal sex differentiation are still largely unclear. The ricefield eel (Monopterus albus), a protogynous hermaphroditic fish with a small genome size (2n = 24), is usually used as an ideal model to study the mechanism of sex differentiation in vertebrates. Therefore, in this study, hsd17b2 gene cDNA was cloned and its mRNA expression profiles were determined in the ricefield eel. The cloned cDNA fragment of hsd17b2 was 1230 bp, including an open reading frame of 1107 bp, encoding 368 amino acid residues with conserved catalytic subunits. Moreover, real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) analysis showed that hsd17b2 mRNA expressed strongly in the ovaries at early developmental stages, weakly in liver and intestine, and barely in testis and other tissues. In particular, hsd17b2 mRNA expression was found to peak in ovaries of young fish and ovotestis at the early stage, and eventually declined in gonads from the late ovotestis to testis. Likewise, chemical in situ hybridization results indicated that the hsd17b2 mRNA signals were primarily detected in the cytoplasm of oogonia and oocytes at stage I–II, subsequently concentrated in the granulosa cells around the oocytes at stage Ⅲ–Ⅳ, but undetectable in mature oocytes and male germ cells. Intriguingly, in ricefield eel ovaries, hsd17b2 mRNA expression could be significantly reduced by 17β-estradiol (E2) or tamoxifen (17β-estradiol inhibitor, E2I) induction at a low concentration (10 ng/mL) and increased by E2I induction at a high concentration (100 ng/mL). On the other hand, both the melatonin (MT) and flutamide (androgen inhibitor, AI) induction could significantly decrease hsd17b2 mRNA expression in the ovary of ricefield eel. This study provides a clue for demonstrating the mechanism of sexual differentiation in fish. The findings of our study imply that the hsd17b2 gene could be a key regulator in sexual differentiation and modulate sex reversal in the ricefield eel and other hermaphroditic fishes. Full article
Show Figures

Figure 1

Figure 1
<p>Identification of <span class="html-italic">hsd17b2</span> cDNA in <span class="html-italic">Monopterus albus.</span> The initiation codon of ATG and stop codon TGA are highlighted in black. The conserved structure of the SDR superfamily, i.e., the Rossmann-folding domain, is highlighted in gray. The classical binding motifs of 17 beta-HSD are in frames, namely TGxxxGxG, YxxxK, PGxxxT, and NNAG. * below the ‘TGA’ for the stop codon.</p>
Full article ">Figure 2
<p>Homologous analysis and phylogenetic tree of Hsd17b2 proteins in <span class="html-italic">Monopterus albus.</span> (<b>A</b>) Multiple alignment of amino acid (aa) sequences of Hsd17b2 proteins. The protein sequences retrieved from NCBI (<a href="https://www.ncbi.nlm.nih.gov/" target="_blank">https://www.ncbi.nlm.nih.gov/</a>, accessed on 16 May 2023), were aligned and calculated based on a ClustalW algorithm in DNAMAN (<a href="https://www.lynnon.com/dnaman.html" target="_blank">https://www.lynnon.com/dnaman.html</a>, accessed on 16 May 2023). The amino acid sequence with 100% identity is highlighted in black, sequences with 75–100% identity are in gray, and sequences with 50% identity are in light gray. The TMD transmembrane region, the Rossmann-folding domain, and low complexity region are highlighted with black frames respectively. The species names and homology are listed at the end of the sequences. (<b>B</b>) Phylogenetic tree of Hsd17b2 proteins. The neighbor-joining phylogenetic tree of Hsd17b2 proteins was constructed by MEGA version 7 (<a href="https://www.megasoftware.net/" target="_blank">https://www.megasoftware.net/</a>, accessed on 16 May 2023), with a set of 1000 bootstraps in the neighbor-joining method. Scale bar tagged with 0.10 indicates the genetic distance, the number on each branch represents the bootstrap value.</p>
Full article ">Figure 3
<p>Bioinformatics analysis of Hsd17b2 protein in <span class="html-italic">Monopterus albus</span>. (<b>A</b>) Ribbon diagram of Hsd17b2 protein, containing α-helices and β-strands. α-helices are shown as coiled ribbons and β-strands as arrows; lines indicate random coils. (<b>B</b>) Predicted transmembrane region; the ordinate represents the probability of transmembrane transfer, and the abscissa indicates the sites of amino acid residues. (<b>C</b>) Hydrophilicity map of Hsd17b2 protein; the ordinate indicates the hydrophilicity index ranging from −2.711 to 2.367. (<b>D</b>) Signal peptide predicted by SignalP 5.0 (<a href="https://services.healthtech.dtu.dk/services/SignalP-5.0/" target="_blank">https://services.healthtech.dtu.dk/services/SignalP-5.0/</a>, accessed on 16 May 2023). The probability of signal protein was about 5.98% and located at residues 1-21.</p>
Full article ">Figure 4
<p>RT-qPCR analysis of <span class="html-italic">zar1</span> and <span class="html-italic">hsd17b2</span> mRNA in tissues. (<b>A</b>,<b>B</b>) Tissue-specific analysis of <span class="html-italic">zar1</span> and <span class="html-italic">hsd17b2</span> mRNA in adult <span class="html-italic">M. albus</span> tissues. A panel of tissues was collected and examined in this study, including the intestine (In), brain (Br), heart (He), liver (Li), spleen (Sp), kidney (Ki), testis (Te), and ovary (Ov). (<b>C</b>) Expression profiles of <span class="html-italic">hsd17b2</span> mRNA in gonads at different developmental stages. (<b>D</b>) Hematoxylin and eosin (HE) staining shows the structures of gonads at different developmental stages examined in this study. Ov1, ovaries from juvenile fish; Ov2, ovaries from young fish; Ov3, ovaries from adult fish; OT1, ovotestis at the early stage; OT2, ovotestis at the middle stage; Te, testis; scale bars, 200 μm. In (<b>A</b>–<b>C</b>), different letters (a–d) represent the significance between groups.</p>
Full article ">Figure 5
<p>Cellular distribution of <span class="html-italic">zar1</span> and <span class="html-italic">hsd17b2</span> mRNA in ovaries. Chemical in situ hybridization was conducted on paraffin-embedded sections of ovaries. Antisense and sense RNA probes of <span class="html-italic">hsd17b2</span> gene were labeled with DIG; signals were developed with NBT/BCIP (in purple). (<b>A</b>–<b>C</b>) Ovaries from intersexual individuals; (<b>D</b>–<b>F</b>) ovaries from juvenile individuals; (<b>G</b>–<b>I</b>) ovaries from young fish. Cellular distribution results indicated that <span class="html-italic">hsd17b2</span> mRNA was strongly expressed in oogonia and then shifted into the somatic cells wrapping up the oocytes filled with yolk. The <span class="html-italic">zar1</span> gene specifically expressed in oocytes was used as the control for analyzing the cellular distribution of <span class="html-italic">hsd17b2</span> mRNA in the gonads of ovotestis at the early stage. I–IV, IIIa, IIIb, represents oocytes at different developmental stages; BV, blood vessels; DO, degenerated oocytes; GL, gonadal lamellae.</p>
Full article ">Figure 6
<p>Cellular distribution of <span class="html-italic">hsd17b2</span> mRNA in ovotestis and testis. (<b>A</b>–<b>C</b>) Ovotestis at the early stage; (<b>D</b>–<b>F</b>) ovotestis at the late stage; (<b>G</b>–<b>I</b>) mature testis. The CISH showed that hsd17b2 mRNA expression patterns in gonads at the ovotestis stages were similar to those in ovaries. However, no signal was detected in the gonads of ovotestis at the late stage, and in testis, the testicular cell nuclei were counterstained by propidium iodide (PI, in red). I–IV, Ⅲa, Ⅲb represent oocytes at the different developmental stages. Sc1, primary spermatocyte; Sc2, secondary spermatocyte; Spd, spermatids; Sp, sperm; BV, blood vessels; DO, degenerated oocytes; GL, gonadal lamellae; MC, mesenchyme cluster.</p>
Full article ">Figure 7
<p>Effects of sex hormone treatment on <span class="html-italic">hsd17b2</span> mRNA expression in the <span class="html-italic">Monopterus albus</span> ovary. The ovaries of young fish were dissected and treated with 17β-estradiol (E2), 17β-estradiol inhibitor (E2I), melatonin (MT), and flutamide (androgen inhibitor, AI) respectively, and <span class="html-italic">hsd17b2</span> mRNA expression in the treated ovaries was examined by RT-qPCR. (<b>A</b>) Treatment with E2 and E2I at 10 ng/mL and 100 ng/mL. (<b>B</b>) MT and AI at 10 ng/mL and 100 ng/mL. The group treated with 0.1% DMSO was set as the control. Each treatment group was set up in triplicates, and the experiment was repeated twice. Data are shown as means ± SEM (n = 3). **, extremely significant (<span class="html-italic">p</span> &lt; 0.01); ns, no significance <span class="html-italic">(p</span> &gt; 0.05).</p>
Full article ">
11 pages, 2766 KiB  
Review
The Emerging Role of Environmental Cadmium Exposure in Prostate Cancer Progression
by Rama Saad, Mohamed Ali Hussein, Gnanasekar Munirathinam and André Kajdacsy-Balla
Environments 2024, 11(8), 181; https://doi.org/10.3390/environments11080181 - 21 Aug 2024
Viewed by 463
Abstract
Cadmium (Cd) is a widespread environmental pollutant with several adverse effects on the general population. While Cd is a well-established risk factor for some cancers, such as lung cancer, its impact on prostate cancer (PCa) is not well understood. PCa mortality is associated [...] Read more.
Cadmium (Cd) is a widespread environmental pollutant with several adverse effects on the general population. While Cd is a well-established risk factor for some cancers, such as lung cancer, its impact on prostate cancer (PCa) is not well understood. PCa mortality is associated with its progression to metastatic spread. This underscores the importance of studying the environmental/or molecular factors that govern the progression from organ-confined tumors to widely metastatic disease. To date, most studies addressing the effects of Cd on PCa are focused on the incidence rather than the progression/outcome. Furthermore, most of these epidemiological studies are limited by the small number of samples and the fact that most of these studies measured Cd levels in the air, blood, or urine, which is less applicable for addressing associations in environmental exposure than the measurement of Cd concentrations in the prostate microenvironment. It is still unknown whether Cd is a driver or a consequence of PCa aggressiveness. Addressing the plausibility of causality requires using proper in vitro and in vivo models for sub-micromolar Cd doses that mimic environmental exposure. Most in vitro studies addressing the functional and molecular effects of Cd are limited by the exclusive use of aggressive PCa cell models and very high micromolar unbound Cd concentrations, which are irrelevant for environmental exposure. Significantly, few studies have addressed the effects of sub-micromolar Cd concentrations. Hence, we suggest using nanomolar concentration that resembles real-life exposure, using less aggressive in vitro models such as RWPE-2, employing 3D organoid culture systems, and adopting high throughput-omics techniques, including metallomics, and using transgenic animal models might represent a more effective model. Here, we focus on reports on the impact of Cd on the progression and aggressiveness of already-established PCa instead of on the initial steps of carcinogenesis. We suggest potential future directions for substantiating the plausible link between Cd exposure and PCa aggressiveness. Full article
Show Figures

Figure 1

Figure 1
<p>Timeline of epidemiological studies [<a href="#B8-environments-11-00181" class="html-bibr">8</a>,<a href="#B20-environments-11-00181" class="html-bibr">20</a>,<a href="#B22-environments-11-00181" class="html-bibr">22</a>,<a href="#B23-environments-11-00181" class="html-bibr">23</a>,<a href="#B24-environments-11-00181" class="html-bibr">24</a>,<a href="#B25-environments-11-00181" class="html-bibr">25</a>,<a href="#B26-environments-11-00181" class="html-bibr">26</a>,<a href="#B27-environments-11-00181" class="html-bibr">27</a>,<a href="#B29-environments-11-00181" class="html-bibr">29</a>,<a href="#B30-environments-11-00181" class="html-bibr">30</a>,<a href="#B31-environments-11-00181" class="html-bibr">31</a>].</p>
Full article ">Figure 2
<p>Cartoon illustration emphasizes the significant role of Cd in promoting PCa progression by activating various signaling pathways. Cd ions permeate the cell through calcium channels and activate critical signaling pathways, including MEK (mitogen-activated protein kinase/extracellular signal-regulated kinase) /ERK1/2 (extracellular signal-regulated kinases 1 and 2), P38 (P38 mitogen-activated protein kinase), JNK (c-Jun N-terminal kinase), and AKT (protein kinase B), via phosphorylation, subsequently leading to increased cell proliferation. Cd also triggers the generation of ROS (reactive oxygen species) in the mitochondria, which in turn induces ER (endoplasmic reticulum) stress. Furthermore, ER stress facilitates the translocation of Smad3 (SMAD family member 3) to the nucleus, resulting in the upregulation of EMT (epithelial–mesenchymal transition) markers such as vimentin and Snail and the downregulation of E-cadherin. This collective action promotes EMT. ↑ denotes an increase in the expression level and ↓ denotes a decrease in the expression level of the corresponding marker.</p>
Full article ">
21 pages, 614 KiB  
Communication
Free Flight Training as a Tool for Psittacine Reintroductions
by Donald J. Brightsmith, Chris Biro, Humberto F. Mendes and Constance Woodman
Birds 2024, 5(3), 522-542; https://doi.org/10.3390/birds5030035 - 21 Aug 2024
Viewed by 1390
Abstract
As habitat loss and other threats accelerate, ecological restoration and reintroduction science are becoming progressively more important. The psittacines are among the most endangered bird groups and are prime candidates for restoration through reintroduction. Unfortunately, post-release survival of captive-raised animals is often quite [...] Read more.
As habitat loss and other threats accelerate, ecological restoration and reintroduction science are becoming progressively more important. The psittacines are among the most endangered bird groups and are prime candidates for restoration through reintroduction. Unfortunately, post-release survival of captive-raised animals is often quite low because, in part, of high predation rates, low site fidelity, poor flight ability, and low flock cohesion. Current best practices in parrot release hold the birds in captivity for a year or more and include distinct methods to address each of these challenges. Here, we conduct a small-scale, proof-of-concept study using free flight methods and human-socialized trained adult birds to hand raise and release a group of six fledgling Blue-and-yellow Macaws in their historical range in southeastern Brazil. All six released birds showed strong flock cohesion and fidelity to the release site, avoided predation, and survived without supplemental feeding for over one year. One bird was captured by local people but was recovered and rereleased and it has reintegrated into the group and is still alive and doing well. The human-socialized trained adult birds modeled both desirable behaviors (flocking, foraging, reacting to predators) and undesirable behaviors and they were returned to captivity before the conclusion of this study. Our study suggests that free flight training has great potential to help captive-raised young attain a broad array of vital skills needed for survival post-release. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Free flight training project process chart. This chart shows the different stages of a release project using free flight training and a human-socialized temporary kernel flock. This manuscript focused on the phases within the dark gray box including the final preparation of the kernel flock, the liberation of the release candidates, and the return of the temporary kernel flock to captivity. However, the full process includes a variety of additional steps including creating or finding a reliable source of fledging age chicks, initial free flight training of the temporary kernel flock, releasing additional birds to the established core release flock, and reproducing the release flock.</p>
Full article ">
Back to TopTop