Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22.014
Filter
1.
Orphanet J Rare Dis ; 19(1): 299, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39148107

ABSTRACT

BACKGROUND: Sirolimus is increasingly utilized in treating diseases associated with mTOR pathway overactivation. Despite its potential, the lack of evidence regarding its long-term safety across all age groups, particularly in pediatric patients, has limited its further application. This study aims to assess the long-term safety of sirolimus, with a specific focus on its impact on growth patterns in pediatric patients. METHODS: This pooled analysis inlcudes two prospective cohort studies spanning 10 years, including 1,738 participants (aged 5 days to 69 years) diagnosed with tuberous sclerosis and/or lymphangioleiomyomatosis. All participants were mTOR inhibitor-naive and received 1 mg/m²/day of sirolimus, with dose adjustments during a two-week titration period to maintain trough blood concentrations between 5 and 10 ng/ml (maximum dose 2 mg). Indicators of physical growth, hematopoietic, liver, renal function, and blood lipid levels were all primary outcomes and were analyzed. The adverse events and related management were also recorded. RESULTS: Sirolimus administration did not lead to deviations from normal growth ranges, but higher doses exhibited a positive association with Z-scores exceeding 2 SD in height, weight, and BMI. Transient elevations in red blood cell and white blood cell counts, along with hyperlipidemia, were primarily observed within the first year of treatment. Other measured parameters remained largely unchanged, displaying only weak correlations with drug use. Stomatitis is the most common adverse event (920/1738, 52.9%). In adult females, menstrual disorders were observed in 48.5% (112/217). CONCLUSIONS: Sirolimus's long-term administration is not associated with adverse effects on children's physical growth pattern, nor significant alterations in hematopoietic, liver, renal function, or lipid levels. A potential dose-dependent influence on growth merits further exploration. TRIAL REGISTRATION: Pediatric patients: Chinese clinical trial registry, No. ChiCTR-OOB-15,006,535. Adult patients: ClinicalTrials, No. NCT03193892.


Subject(s)
Sirolimus , Humans , Sirolimus/adverse effects , Sirolimus/therapeutic use , Child , Female , Adolescent , Child, Preschool , Adult , Male , Infant , Young Adult , Middle Aged , Infant, Newborn , Aged , Tuberous Sclerosis/drug therapy , Lymphangioleiomyomatosis/drug therapy , Prospective Studies
2.
Methods Mol Biol ; 2818: 213-226, 2024.
Article in English | MEDLINE | ID: mdl-39126477

ABSTRACT

Conditional depletion of proteins is a potential strategy to elucidate protein function, especially in complex cellular processes like meiosis. Several methods are available to effectively deplete a protein in a conditional manner. Conditional loss of a protein function can be achieved by depleting it from its region of action by degrading it. A conditional loss of protein function can also be achieved by sequestering it to a functionally unavailable compartment inside the cell. This chapter describes anchor away, a conditional depletion tool that can deplete a protein both temporally and spatially by translocation. It utilizes the affinity of FRB to bind FKBP12 in the presence of rapamycin for a quick and efficient translocation of the protein to a designated location. Anchor away is a reliable tool for the study of meiotic proteins, as only small quantities of rapamycin are required to efficiently and rapidly translocate the protein of interest without compromising meiotic progression.


Subject(s)
Meiosis , Protein Transport , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Sirolimus , Sirolimus/pharmacology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Nuclear Proteins/metabolism , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Protein 1A/genetics
3.
Commun Biol ; 7(1): 974, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127848

ABSTRACT

Calorie restriction (CR) and treatment with rapamycin (RM), an inhibitor of the mTORC1 growth-promoting signaling pathway, are known to slow aging and promote health from worms to humans. At the transcriptome and proteome levels, long-term CR and RM treatments have partially overlapping effects, while their impact on protein phosphorylation within cellular signaling pathways have not been compared. Here we measured the phosphoproteomes of soleus, tibialis anterior, triceps brachii and gastrocnemius muscles from adult (10 months) and 30-month-old (aged) mice receiving either a control, a calorie restricted or an RM containing diet from 15 months of age. We reproducibly detected and extensively analyzed a total of 6960 phosphosites, 1415 of which are not represented in standard repositories. We reveal the effect of these interventions on known mTORC1 pathway substrates, with CR displaying greater between-muscle variation than RM. Overall, CR and RM have largely consistent, but quantitatively distinct long-term effects on the phosphoproteome, mitigating age-related changes to different degrees. Our data expands the catalog of protein phosphorylation sites in the mouse, providing important information regarding their tissue-specificity, and revealing the impact of long-term nutrient-sensing pathway inhibition on mouse skeletal muscle.


Subject(s)
Aging , Caloric Restriction , Muscle, Skeletal , Sirolimus , Animals , Phosphorylation , Aging/metabolism , Sirolimus/pharmacology , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Male , Mice, Inbred C57BL , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction/drug effects , Proteome/metabolism , Phosphoproteins/metabolism , Phosphoproteins/genetics , Muscle Proteins/metabolism
4.
Theranostics ; 14(11): 4375-4392, 2024.
Article in English | MEDLINE | ID: mdl-39113803

ABSTRACT

Rationale: Autism spectrum disorder (ASD) represents a complex neurodevelopmental condition lacking specific pharmacological interventions. Given the multifaced etiology of ASD, there exist no effective treatment for ASD. Rapamycin (RAPA) can activate autophagy by inhibiting the mTOR pathway and has exhibited promising effects in treating central nervous system disorders; however, its limited ability to cross the blood-brain barrier (BBB) has hindered its clinical efficacy, leading to substantial side effects. Methods: To address this challenge, we designed a drug delivery system utilizing red blood cell membrane (CM) vesicles modified with SS31 peptides to enhance the brain penetration of RAPA for the treatment of autism. Results: The fabricated SCM@RAPA nanoparticles, with an average diameter of 110 nm, exhibit rapid release of RAPA in a pathological environment characterized by oxidative stress. In vitro results demonstrate that SCM@RAPA effectively activate cellular autophagy, reduce intracellular ROS levels, improve mitochondrial function, thereby ameliorating neuronal damage. SS31 peptide modification significantly enhances the BBB penetration and rapid brain accumulation of SCM@RAPA. Notably, SCM@RAPA nanoparticles demonstrate the potential to ameliorate social deficits, improve cognitive function, and reverse neuronal impairments in valproic acid (VPA)-induced ASD models. Conclusions: The therapeutic potential of SCM@RAPA in managing ASD signifies a paradigm shift in autism drug treatment, holding promise for clinical interventions in diverse neurological conditions.


Subject(s)
Autism Spectrum Disorder , Autophagy , Blood-Brain Barrier , Nanoparticles , Oxidative Stress , Sirolimus , Sirolimus/administration & dosage , Sirolimus/pharmacology , Oxidative Stress/drug effects , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/metabolism , Animals , Autophagy/drug effects , Nanoparticles/chemistry , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Mice , Humans , Drug Delivery Systems/methods , Disease Models, Animal , Male , Biomimetic Materials/administration & dosage , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Biomimetics/methods , Brain/metabolism , Brain/drug effects , Peptides/administration & dosage , Reactive Oxygen Species/metabolism , Valproic Acid/administration & dosage , Valproic Acid/pharmacology
5.
Int J Nanomedicine ; 19: 7871-7893, 2024.
Article in English | MEDLINE | ID: mdl-39114180

ABSTRACT

Purpose: Ovarian cancer has the highest mortality rate and lowest survival rate among female reproductive system malignancies. There are treatment options of surgery and chemotherapy, but both are limited. In this study, we developed and evaluated micelles composed of D-α-tocopheryl polyethylene-glycol (PEG) 1000 succinate (TPGS) and Soluplus® (SOL) loaded with olaparib (OLA), a poly(ADP-ribose)polymerase (PARP) inhibitor, and rapamycin (RAPA), a mammalian target of rapamycin (mTOR) inhibitor in ovarian cancer. Methods: We prepared micelles containing different molar ratios of OLA and RAPA embedded in different weight ratios of TPGS and SOL (OLA/RAPA-TPGS/SOL) were prepared and physicochemical characterized. Furthermore, we performed in vitro cytotoxicity experiments of OLA, RAPA, and OLA/RAPA-TPGS/SOL. In vivo toxicity and antitumor efficacy assays were also performed to assess the efficacy of the mixed micellar system. Results: OLA/RAPA-TPGS/SOL containing a 4:1 TPGS:SOL weight ratio and a 2:3 OLA:RAPA molar ratio showed synergistic effects and were optimized. The drug encapsulation efficiency of this formulation was >65%, and the physicochemical properties were sustained for 180 days. Moreover, the formulation had a high cell uptake rate and significantly inhibited cell migration (**p < 0.01). In the in vivo toxicity test, no toxicity was observed, with the exception of the high dose group. Furthermore, OLA/RAPA-TPGS/SOL markedly inhibited tumor spheroid and tumor growth in vivo. Conclusion: Compared to the control, OLA/RAPA-TPGS/SOL showed significant tumor inhibition. These findings lay a foundation for the use of TPGS/SOL mixed micelles loaded with OLA and RAPA in the treatment of ovarian cancer.


Subject(s)
Micelles , Ovarian Neoplasms , Phthalazines , Piperazines , Polyethylene Glycols , Polyvinyls , Sirolimus , Vitamin E , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Piperazines/chemistry , Piperazines/pharmacology , Polyethylene Glycols/chemistry , Humans , Animals , Cell Line, Tumor , Vitamin E/chemistry , Vitamin E/pharmacology , Sirolimus/chemistry , Sirolimus/pharmacology , Sirolimus/administration & dosage , Sirolimus/pharmacokinetics , Phthalazines/chemistry , Phthalazines/pharmacology , Phthalazines/administration & dosage , Phthalazines/pharmacokinetics , Polyvinyls/chemistry , Polyvinyls/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Mice , Drug Carriers/chemistry , Xenograft Model Antitumor Assays , Mice, Nude , Mice, Inbred BALB C , Cell Survival/drug effects
6.
J Nanobiotechnology ; 22(1): 486, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143545

ABSTRACT

Lower back pain (LBP) is a common condition closely associated with intervertebral disc degeneration (IDD), causing a significant socioeconomic burden. Inflammatory activation in degenerated discs involves pro-inflammatory cytokines, dysregulated regulatory cytokines, and increased levels of nerve growth factor (NGF), leading to further intervertebral disc destruction and pain sensitization. Macrophage polarization is closely related to autophagy. Based on these pathological features, a structured biomimetic nanoparticle coated with TrkA-overexpressing macrophage membranes (TMNP@SR) with a rapamycin-loaded mesoporous silica core is developed. TMNP@SR acted like sponges to adsorbe inflammatory cytokines and NGF and delivers the autophagy regulator rapamycin (RAPA) into macrophages through homologous targeting effects of the outer engineered cell membrane. By regulating autophagy activation, TMNP@SR promoted the M1-to-M2 switch of macrophages to avoid continuous activation of inflammation within the degenerated disc, which prevented the apoptosis of nucleus pulposus cells. In addition, TMNP@SR relieved mechanical and thermal hyperalgesia, reduced calcitonin gene-related peptide (CGRP) and substance P (SP) expression in the dorsal root ganglion, and downregulated GFAP and c-FOS signaling in the spinal cord in the rat IDD model. In summary, TMNP@SR spontaneously inhibits the aggravation of disc inflammation to alleviate disc degeneration and reduce the ingress of sensory nerves, presenting a promising treatment strategy for LBP induced by disc degeneration.


Subject(s)
Autophagy , Intervertebral Disc Degeneration , Nanoparticles , Rats, Sprague-Dawley , Intervertebral Disc Degeneration/drug therapy , Intervertebral Disc Degeneration/metabolism , Animals , Autophagy/drug effects , Nanoparticles/chemistry , Rats , Male , Mice , Macrophages/drug effects , Macrophages/metabolism , Low Back Pain/drug therapy , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Sirolimus/pharmacology , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology , Nucleus Pulposus/metabolism , Inflammation/drug therapy , Cytokines/metabolism , Biomimetics/methods , Disease Models, Animal , Nerve Growth Factor/metabolism , RAW 264.7 Cells
7.
Nat Commun ; 15(1): 6485, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090122

ABSTRACT

The modular nature of polyketide assembly lines and the significance of their products make them prime targets for combinatorial engineering. The recently updated module boundary has been successful for engineering short synthases, yet larger synthases constructed using the updated boundary have not been investigated. Here we describe our design and implementation of a BioBricks-like platform to rapidly construct 5 triketide, 25 tetraketide, and 125 pentaketide synthases to test every module combination of the pikromycin synthase. Anticipated products are detected from 60% of the triketide synthases, 32% of the tetraketide synthases, and 6.4% of the pentaketide synthases. We determine ketosynthase gatekeeping and module-skipping are the principal impediments to obtaining functional synthases. The platform is also employed to construct active hybrid synthases by incorporating modules from the erythromycin, spinosyn, and rapamycin assembly lines. The relaxed gatekeeping of a ketosynthase in the rapamycin synthase is especially encouraging in the quest to produce designer polyketides.


Subject(s)
Macrolides , Polyketide Synthases , Polyketide Synthases/metabolism , Polyketide Synthases/genetics , Macrolides/metabolism , Protein Engineering/methods , Erythromycin , Polyketides/metabolism , Polyketides/chemistry , Streptomyces/enzymology , Streptomyces/genetics , Sirolimus , Bacterial Proteins/metabolism , Bacterial Proteins/genetics
10.
BMC Genomics ; 25(1): 766, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39107687

ABSTRACT

BACKGROUND: Many common diseases exhibit uncontrolled mTOR signaling, prompting considerable interest in the therapeutic potential of mTOR inhibitors, such as rapamycin, to treat a range of conditions, including cancer, aging-related pathologies, and neurological disorders. Despite encouraging preclinical results, the success of mTOR interventions in the clinic has been limited by off-target side effects and dose-limiting toxicities. Improving clinical efficacy and mitigating side effects require a better understanding of the influence of key clinical factors, such as sex, tissue, and genomic background, on the outcomes of mTOR-targeting therapies. RESULTS: We assayed gene expression with and without rapamycin exposure across three distinct body parts (head, thorax, abdomen) of D. melanogaster flies, bearing either their native melanogaster mitochondrial genome or the mitochondrial genome from a related species, D. simulans. The fully factorial RNA-seq study design revealed a large number of genes that responded to the rapamycin treatment in a sex-dependent and tissue-dependent manner, and relatively few genes with the transcriptional response to rapamycin affected by the mitochondrial background. Reanalysis of an earlier study confirmed that mitochondria can have a temporal influence on rapamycin response. CONCLUSIONS: We found significant and wide-ranging effects of sex and body part, alongside a subtle, potentially time-dependent, influence of mitochondria on the transcriptional response to rapamycin. Our findings suggest a number of pathways that could be crucial for predicting potential side effects of mTOR inhibition in a particular sex or tissue. Further studies of the temporal response to rapamycin are necessary to elucidate the effects of the mitochondrial background on mTOR and its inhibition.


Subject(s)
Mitochondria , Sirolimus , Animals , Sirolimus/pharmacology , Female , Male , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/drug effects , Sex Factors , TOR Serine-Threonine Kinases/metabolism , Organ Specificity/genetics , Drosophila/genetics , Drosophila/drug effects , Transcription, Genetic/drug effects , Gene Expression Profiling
11.
Int J Mol Med ; 54(4)2024 Oct.
Article in English | MEDLINE | ID: mdl-39129316

ABSTRACT

Topical therapy remains a critical component in the management of immune­mediated inflammatory dermatoses such as psoriasis and atopic dermatitis. In this field, macrolactam immunomodulators, including calcineurin and mammalian target of rapamycin inhibitors, can offer steroid­free therapeutic alternatives. Despite their potential for skin­selective treatment compared with topical corticosteroids, the physicochemical properties of these compounds, such as high lipophilicity and large molecular size, do not meet the criteria for efficient penetration into the skin, especially with conventional topical vehicles. Thus, more sophisticated approaches are needed to address the pharmacokinetic limitations of traditional formulations. In this regard, interest has increasingly focused on nanoparticulate systems to optimize penetration kinetics and enhance the efficacy and safety of topical calcineurin and mTOR inhibitors in inflamed skin. Several types of nanovectors have been explored as topical carriers to deliver tacrolimus in both psoriatic and atopic skin, while preclinical data on nanocarrier­based delivery of topical sirolimus in inflamed skin are also emerging. Given the promising preliminary outcomes and the complexities of drug delivery across inflamed skin, further research is required to translate these nanotherapeutics into clinical settings for inflammatory skin diseases. The present review outlined the dermatokinetic profiles of topical calcineurin and mTOR inhibitors, particularly tacrolimus, pimecrolimus and sirolimus, focusing on their penetration kinetics in psoriatic and atopic skin. It also summarizes the potential anti­inflammatory benefits of topical sirolimus and explores novel preclinical studies investigating dermally applied nanovehicles to evaluate and optimize the skin delivery, efficacy and safety of these 'hard­to­formulate' macromolecules in the context of psoriasis and atopic dermatitis.


Subject(s)
Calcineurin Inhibitors , MTOR Inhibitors , Humans , Calcineurin Inhibitors/therapeutic use , Animals , MTOR Inhibitors/therapeutic use , Administration, Topical , Nanotechnology/methods , Dermatitis, Atopic/drug therapy , Nanoparticles/chemistry , Tacrolimus/therapeutic use , Tacrolimus/administration & dosage , Tacrolimus/pharmacology , Sirolimus/therapeutic use , Sirolimus/pharmacology , Drug Delivery Systems/methods
12.
Clin Transl Sci ; 17(8): e70014, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39162578

ABSTRACT

Dose optimization of sirolimus may further improve outcomes in allogeneic hematopoietic cell transplant (HCT) patients receiving post-transplantation cyclophosphamide (PTCy) to prevent graft-versus-host disease (GVHD). Sirolimus exposure-response association studies in HCT patients (i.e., the association of trough concentration with clinical outcomes) have been conflicting. Sirolimus has important effects on T-cells, including conventional (Tcons) and regulatory T-cells (Tregs), both of which have been implicated in the mechanisms by which PTCy prevents GVHD, but there is an absence of validated biomarkers of sirolimus effects on these cell subsets. Considering the paucity of existing biomarkers and the complexities of the immune system, we conducted a literature review to inform a quantitative systems pharmacology (QSP) model of GVHD. The published literature presented multiple challenges. The sirolimus pharmacokinetic models insufficiently describe sirolimus distribution to relevant physiological compartments. Despite multiple publications describing sirolimus effects on Tcons and Tregs in preclinical and human ex vivo models, consistent parameters relating sirolimus concentrations to circulating Tcons and Tregs could not be found. Each aspect presents a challenge in building a QSP model of sirolimus and its temporal effects on T-cell subsets and GVHD prevention. To optimize GVHD prevention regimens, phase I studies and systematic studies of immunosuppressant concentration-effect association are needed for QSP modeling.


Subject(s)
Cyclophosphamide , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents , Sirolimus , Humans , Sirolimus/administration & dosage , Graft vs Host Disease/prevention & control , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Cyclophosphamide/adverse effects , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacokinetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Animals , Models, Biological
13.
J Pharmacol Sci ; 156(1): 9-18, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39068035

ABSTRACT

Necroptosis, necrosis characterized by RIPK3-MLKL activation, has been proposed as a mechanism of doxorubicin (DOX)-induced cardiomyopathy. We showed that rapamycin, an mTORC1 inhibitor, attenuates cardiomyocyte necroptosis. Here we examined role of MLKL in DOX-induced myocardial damage and protective effects of rapamycin. Cardiomyopathy was induced in mice by intraperitoneal injections of DOX (10 mg/kg, every other day) and followed for 7 days. DOX-treated mice showed a significant decline in LVEF assessed by cardiac MRI (45.5 ± 5.1% vs. 65.4 ± 4.2%), reduction in overall survival rates, and increases in myocardial RIPK3 and MLKL expression compared with those in vehicle-treated mice, and those changes were prevented by administration of rapamycin (0.25 mg/kg) before DOX injection. In immunohistochemical analyses, p-MLKL signals were detected in the cardiomyocytes of DOX-treated mice, and the signals were reduced by rapamycin. Mlkl+/- and Mlkl-/- mice were similarly resistant to DOX-induced cardiac dysfunction, indicating that a modest reduction in MLKL level is sufficient to prevent the development of DOX-induced cardiomyopathy. However, evidence of cardiomyocyte necrosis assessed by C9 immunostaining, presence of replacement fibrosis, and electron microscopic analyses was negligible in the myocardium of DOX-treated mice. Thus, MLKL-mediated signaling contributes to DOX-induced cardiac dysfunction primarily by a necrosis-independent mechanism, which is inhibitable by rapamycin.


Subject(s)
Cardiomyopathies , Doxorubicin , Mice, Inbred C57BL , Myocytes, Cardiac , Necroptosis , Protein Kinases , Receptor-Interacting Protein Serine-Threonine Kinases , Sirolimus , Animals , Doxorubicin/adverse effects , Protein Kinases/metabolism , Sirolimus/pharmacology , Cardiomyopathies/chemically induced , Cardiomyopathies/prevention & control , Cardiomyopathies/pathology , Cardiomyopathies/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Necroptosis/drug effects , Male , Mice , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/toxicity
14.
Xenotransplantation ; 31(4): e12876, 2024.
Article in English | MEDLINE | ID: mdl-39031102

ABSTRACT

BACKGROUND: Long-term immunosuppressive maintenance therapy is necessary to prevent the rejection of xenografts. However, it is still unclear which oral immunosuppressant is most suitable for pig-to-human xenotransplantation . METHODS: A xenogeneic mixed lymphocyte reaction (MLR) system was established using peripheral blood mononuclear cells (PBMCs) isolated from wildtype (WT) or GTKO/CMAHKO/ß4GalNT2KO (TKO) pigs as stimulator cells and human PBMCs as responder cells. Various concentrations of tacrolimus (Tac), cyclosporine (CsA), or rapamycin (Rapa) were added to the MLR system as interventions. The inhibitory effects of the three immunosuppressants on the proliferation and cytokine production of human T cells were studied and compared. The inhibitory effect of anti-CD154 mAb alone or in combination with Tac/CsA/Rapa on xenoreactive MLR was also investigated. RESULTS: PBMCs from both WT and TKO pigs stimulated significant proliferation of human T cells. Tac had a strong inhibitory effect on human T-cell proliferation stimulated by pig PBMCs. CsA inhibited human T-cell proliferation in a typical dose-dependent manner. When Tac and CsA concentrations reached 5 and 200 ng/mL, respectively, the proliferation rates of CD3+/CD4+/CD8+ T cells were reduced almost to a negative level. Even at high concentrations, Rapa had only a moderate inhibitory effect on xenogeneic MLR. The inhibitory effects of these three immunosuppressants on xenogeneic T-cell responses were further confirmed by the detection of CD25 expression and supernatant cytokines (IL-2, IL-6, IFN-γ, TNF-α, IL-4, IL-10, and IL-17). Although anti-CD154 mAb monotherapy showed only moderate inhibitory effects on xenoreactive T-cell proliferation, low-dose anti-CD154 mAb combined with low-dose Tac, CSA, or Rapa could produce significant synergistic inhibitory effects. CONCLUSION: Tac is more efficient than CsA or Rapa in inhibiting xenogeneic T-cell responses in vitro. If used in combination with anti-CD154 mAb, all the three immunosuppressants can achieve satisfactory synergistic inhibitory effects.


Subject(s)
Cell Proliferation , Cyclosporine , Immunosuppressive Agents , Lymphocyte Culture Test, Mixed , Sirolimus , Tacrolimus , Transplantation, Heterologous , Animals , Sirolimus/pharmacology , Humans , Tacrolimus/pharmacology , Immunosuppressive Agents/pharmacology , Cyclosporine/pharmacology , Transplantation, Heterologous/methods , Swine , Cell Proliferation/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Graft Rejection/immunology , Graft Rejection/prevention & control , Cytokines/metabolism , Cytokines/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/drug effects , Animals, Genetically Modified
15.
Eur J Pharm Sci ; 200: 106837, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38960206

ABSTRACT

Intractable lymphatic malformations (iLM) pose a significant threat to affected children, demonstrating limited responses to conventional treatments. Sirolimus, effectively inhibiting endothelial cell proliferation in lymphatic vessels, plays a crucial role in iLM treatment. However, the drug's narrow therapeutic window and substantial interindividual variability necessitate customized dosing strategies. This study aims to establish a Population Pharmacokinetic Model (PopPK model) for sirolimus in pediatric iLM patients, identifying quantitative relationships between covariates and sirolimus clearance and volume of distribution. Initial dosages are recommended based on a target concentration range of 5-15 ng/mL. Retrospective data from our institution, encompassing 53 pediatric patients with 275 blood concentration results over the past five years (average age: 4.64 ± 4.19 years), constituted the foundation of this analysis. The final model, adopting a first-order absorption and elimination single-compartment model, retained age as the sole covariate. Results indicated a robust correlation between apparent clearance (CL/F) at 5.56 L/h, apparent volume of distribution (V/F) at 292.57 L, and age. Monte Carlo simulation guided initial dosages for patients aged 0-18 years within the target concentration range. This study presents the first PopPK model using a large Therapeutic Drug Monitoring (TDM) database to describe personalized sirolimus dosing for pediatric iLM patients, contributing to pharmacokinetic guidance and potentially improving long-term clinical outcomes.


Subject(s)
Lymphatic Abnormalities , Models, Biological , Sirolimus , Humans , Sirolimus/pharmacokinetics , Sirolimus/administration & dosage , Sirolimus/blood , Child , Child, Preschool , Female , Male , Infant , Adolescent , Lymphatic Abnormalities/drug therapy , Retrospective Studies , Monte Carlo Method , Infant, Newborn , Precision Medicine/methods , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/blood
16.
Sci Rep ; 14(1): 15140, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956234

ABSTRACT

Rapamycin slows cystogenesis in murine models of polycystic kidney disease (PKD) but failed in clinical trials, potentially due to insufficient drug dosing. To improve drug efficiency without increasing dose, kidney-specific drug delivery may be used. Mesoscale nanoparticles (MNP) selectively target the proximal tubules in rodents. We explored whether MNPs can target cystic kidney tubules and whether rapamycin-encapsulated-MNPs (RapaMNPs) can slow cyst growth in Pkd1 knockout (KO) mice. MNP was intravenously administered in adult Pkd1KO mice. Serum and organs were harvested after 8, 24, 48 or 72 h to measure MNP localization, mTOR levels, and rapamycin concentration. Pkd1KO mice were then injected bi-weekly for 6 weeks with RapaMNP, rapamycin, or vehicle to determine drug efficacy on kidney cyst growth. Single MNP injections lead to kidney-preferential accumulation over other organs, specifically in tubules and cysts. Likewise, one RapaMNP injection resulted in higher drug delivery to the kidney compared to the liver, and displayed sustained mTOR inhibition. Bi-weekly injections with RapaMNP, rapamycin or vehicle for 6 weeks resulted in inconsistent mTOR inhibition and little change in cyst index, however. MNPs serve as an effective short-term, kidney-specific delivery system, but long-term RapaMNP failed to slow cyst progression in Pkd1KO mice.


Subject(s)
Disease Models, Animal , Mice, Knockout , Nanoparticles , Polycystic Kidney Diseases , Sirolimus , Animals , Sirolimus/administration & dosage , Sirolimus/pharmacology , Mice , Polycystic Kidney Diseases/drug therapy , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/pathology , Nanoparticles/administration & dosage , TOR Serine-Threonine Kinases/metabolism , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , Kidney/metabolism , Kidney/drug effects , Kidney/pathology , Drug Delivery Systems , Male
17.
Genome Med ; 16(1): 85, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956711

ABSTRACT

BACKGROUND: Restraining or slowing ageing hallmarks at the cellular level have been proposed as a route to increased organismal lifespan and healthspan. Consequently, there is great interest in anti-ageing drug discovery. However, this currently requires laborious and lengthy longevity analysis. Here, we present a novel screening readout for the expedited discovery of compounds that restrain ageing of cell populations in vitro and enable extension of in vivo lifespan. METHODS: Using Illumina methylation arrays, we monitored DNA methylation changes accompanying long-term passaging of adult primary human cells in culture. This enabled us to develop, test, and validate the CellPopAge Clock, an epigenetic clock with underlying algorithm, unique among existing epigenetic clocks for its design to detect anti-ageing compounds in vitro. Additionally, we measured markers of senescence and performed longevity experiments in vivo in Drosophila, to further validate our approach to discover novel anti-ageing compounds. Finally, we bench mark our epigenetic clock with other available epigenetic clocks to consolidate its usefulness and specialisation for primary cells in culture. RESULTS: We developed a novel epigenetic clock, the CellPopAge Clock, to accurately monitor the age of a population of adult human primary cells. We find that the CellPopAge Clock can detect decelerated passage-based ageing of human primary cells treated with rapamycin or trametinib, well-established longevity drugs. We then utilise the CellPopAge Clock as a screening tool for the identification of compounds which decelerate ageing of cell populations, uncovering novel anti-ageing drugs, torin2 and dactolisib (BEZ-235). We demonstrate that delayed epigenetic ageing in human primary cells treated with anti-ageing compounds is accompanied by a reduction in senescence and ageing biomarkers. Finally, we extend our screening platform in vivo by taking advantage of a specially formulated holidic medium for increased drug bioavailability in Drosophila. We show that the novel anti-ageing drugs, torin2 and dactolisib (BEZ-235), increase longevity in vivo. CONCLUSIONS: Our method expands the scope of CpG methylation profiling to accurately and rapidly detecting anti-ageing potential of drugs using human cells in vitro, and in vivo, providing a novel accelerated discovery platform to test sought after anti-ageing compounds and geroprotectors.


Subject(s)
Aging , DNA Methylation , Longevity , Humans , Animals , DNA Methylation/drug effects , Longevity/drug effects , Aging/drug effects , Epigenesis, Genetic/drug effects , Drug Discovery/methods , Cellular Senescence/drug effects , Drug Evaluation, Preclinical/methods , Drosophila , Cells, Cultured , Sirolimus/pharmacology
19.
J Nanobiotechnology ; 22(1): 417, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39014353

ABSTRACT

Sirolimus (SR) is a macrolide with antifungal and antitumor immunosuppressant properties, classified as a selective inhibitor of mammalian target of rapamycin (mTOR). In this study, an ionic in situ gel of SR (SR-SUS-ISG) was formulated using gellan gum, exhibiting stability regardless of temperature and pH variations, causing minimal irritation. Harnessing the physiological conditions of the eye, SR-SUS-ISG underwent gelation upon contact with ions, increasing drug viscosity and prolonging retention on the ocular surface. Concurrently, SR-SUS-ISG displayed favorable shear dilution properties, reducing viscosity at ambient temperature, enhancing fluidity, and facilitating convenient packaging and transport. Biocompatibility assessments on both human corneal epithelial cells and rabbit eyes demonstrated that SR-SUS-ISG could well be tolerated. Pharmacokinetic investigations in rabbit ocular aqueous humor revealed sustained release, improved corneal penetration, and enhanced bioavailability. Additionally, in a rat corneal alkali burn model, SR-SUS-ISG exhibited inhibitory effects on corneal neovascularization, associated with decreased levels of the inflammatory factors VEGF and MMPs. These findings suggested that SR-SUS-ISG held promise as an effective ocular drug delivery system.


Subject(s)
Gels , Sirolimus , Animals , Rabbits , Sirolimus/pharmacology , Sirolimus/pharmacokinetics , Sirolimus/chemistry , Humans , Gels/chemistry , Cornea/drug effects , Cornea/metabolism , Rats , Male , Polysaccharides, Bacterial/chemistry , Nanoparticles/chemistry , Administration, Ophthalmic , Corneal Neovascularization/drug therapy , Rats, Sprague-Dawley , Viscosity , Drug Delivery Systems , Ophthalmic Solutions/chemistry , Ophthalmic Solutions/pharmacology , Cell Line , Biological Availability
20.
J Neuroimmune Pharmacol ; 19(1): 40, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39078442

ABSTRACT

The brain and immune system communicate through complex bidirectional pathways, but the specificity by which the brain perceives or even remembers alterations in immune homeostasis is still poorly understood. Recent data revealed that immune-related information under peripheral inflammatory conditions, termed as "immunengram", were represented in specific neuronal ensembles in the insular cortex (IC). Chemogenetic reactivation of these neuronal ensembles was sufficient to retrieve the inflammatory stages, indicating that the brain can store and retrieve specific immune responses. Against this background, the current approach was designed to investigate the ability of the IC to process states of immunosuppression pharmacologically induced by the mechanistic target of rapamycin (mTOR) inhibitor rapamycin. We here show that the IC perceives the initial state of immunosuppression, reflected by increased deep-brain electroencephalography (EEG) activity during acute immunosuppressive drug treatment. Following an experienced period of immunosuppression, though, diminished splenic cytokine production as formerly induced by rapamycin could not be reinstated by nonspecific chemogenetic activation or inhibition of the IC. These findings suggest that the information of a past, or experienced status of pharmacologically induced immunosuppression is not represented in the IC. Together, the present work extends the view of immune-to-brain communication during the states of peripheral immunosuppression and foster the prominent role of the IC for interoception.


Subject(s)
Immunosuppressive Agents , Insular Cortex , Sirolimus , Animals , Sirolimus/pharmacology , Insular Cortex/drug effects , Male , Immunosuppressive Agents/pharmacology , Electroencephalography , Immunosuppression Therapy/methods , Cytokines/metabolism , Cytokines/immunology , Mice , Immune Tolerance/drug effects , Immune Tolerance/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/immunology , Cerebral Cortex/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL