Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 18.419
Filter
1.
Hand Clin ; 40(3): 429-440, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38972687

ABSTRACT

This article highlights the use of rodents as preclinical models to evaluate the management of nerve injuries, describing the pitfalls and value from rodent nerve injury and regeneration outcomes, as well as treatments derived from these rodent models. The anatomic structure, size, and cellular and molecular differences and similarities between rodent and human nerves are summarized. Specific examples of success and failure when assessing outcome metrics are presented for context. Evidence for translation to clinical practice includes the topics of electrical stimulation, Tacrolimus (FK506), and acellular nerve allografts.


Subject(s)
Disease Models, Animal , Nerve Regeneration , Peripheral Nerve Injuries , Animals , Peripheral Nerve Injuries/surgery , Peripheral Nerve Injuries/therapy , Nerve Regeneration/physiology , Rats , Translational Research, Biomedical , Humans , Tacrolimus , Rodentia , Electric Stimulation Therapy , Immunosuppressive Agents , Mice
4.
Sci Rep ; 14(1): 13303, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38858399

ABSTRACT

Dengue virus (DENV) infection is a public health concern in several countries and is associated with severe diseases, such as dengue hemorrhagic fever and dengue shock syndrome. DENVs are transmitted to humans via the bites of infected Aedes mosquitoes, and no antiviral therapeutics are currently available. In this work, we aimed to identify antiviral drugs against DENV type 2 (DENV2) infections and selected pimecrolimus as a potential antiviral drug candidate. Pimecrolimus significantly inhibited DENV2-mediated cell death and replication in vitro. We also confirmed a decrease in the number of plaques formed as well as in the envelope protein levels of DENV2. The time-of-addition and course experiments revealed that pimecrolimus inhibited DENV2 infection during the early stages of the virus replication cycle. In an experimental mouse model, orally administered pimecrolimus alleviated body weight loss and lethality caused by DENV2 infection, which we used as readouts of the drug's antiviral potency. Furthermore, pimecrolimus significantly inhibited the DENV2 load and ameliorated focal necrosis in the liver and spleen. Taken together, our in vitro and in vivo findings suggest that pimecrolimus is a promising antiviral drug candidate for the treatment of DENV2 infection.


Subject(s)
Antiviral Agents , Dengue Virus , Dengue , Tacrolimus , Virus Replication , Animals , Dengue Virus/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Tacrolimus/therapeutic use , Virus Replication/drug effects , Mice , Dengue/drug therapy , Dengue/virology , Humans , Disease Models, Animal , Chlorocebus aethiops , Cell Line , Vero Cells
5.
BMJ Open ; 14(6): e086777, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871657

ABSTRACT

INTRODUCTION: Most solid organ transplants originate from donors meeting criteria for death by neurological criteria (DNC). Within the organ donor, physiological responses to brain death increase the risk of ischaemia reperfusion injury and delayed graft function. Donor preconditioning with calcineurin inhibition may reduce this risk. METHODS AND ANALYSIS: We designed a multicentre placebo-controlled pilot randomised trial involving nine organ donation hospitals and all 28 transplant programmes in the Canadian provinces of Ontario and Québec. We planned to enrol 90 DNC donors and their approximately 324 organ recipients, totalling 414 participants. Donors receive an intravenous infusion of either tacrolimus 0.02 mg/kg over 4 hours prior to organ retrieval, or a matching placebo, while monitored in an intensive care unit for any haemodynamic changes during the infusion. Among all study organ recipients, we record measures of graft function for the first 7 days in hospital and we will record graft survival after 1 year. We examine the feasibility of this trial with respect to the proportion of all eligible donors enrolled and the proportion of all eligible transplant recipients consenting to receive a CINERGY organ transplant and to allow the use of their health data for study purposes. We will report these feasibility outcomes as proportions with 95% CIs. We also record any barriers encountered in the launch and in the implementation of this trial with detailed source documentation. ETHICS AND DISSEMINATION: We will disseminate trial results through publications and presentations at participating sites and conferences. This study has been approved by Health Canada (HC6-24-c241083) and by the Research Ethics Boards of all participating sites and in Québec (MP-31-2020-3348) and Clinical Trials Ontario (Project #3309). TRIAL REGISTRATION NUMBER: NCT05148715.


Subject(s)
Calcineurin Inhibitors , Delayed Graft Function , Kidney Transplantation , Tissue Donors , Humans , Calcineurin Inhibitors/administration & dosage , Calcineurin Inhibitors/therapeutic use , Pilot Projects , Delayed Graft Function/prevention & control , Tacrolimus/therapeutic use , Tacrolimus/administration & dosage , Brain Death , Graft Survival/drug effects , Quebec , Randomized Controlled Trials as Topic , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Multicenter Studies as Topic , Male , Ontario , Adult , Female
6.
Clin Transl Sci ; 17(6): e13860, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38923308

ABSTRACT

The recommended immunosuppressive treatment after kidney transplantation consists of tacrolimus, mycophenolate mofetil, and low-dose corticosteroids. Drug concentrations are monitored using therapeutic drug monitoring (TDM), which does not necessarily correlate with pharmacodynamic activity. To find the balance between optimal efficacy and minimal toxicity, it might be more informative to monitor patients' immunological status rather than drug concentrations. We selected a panel of T-cell-based immune assays, which were used for immunomonitoring of 14 stable kidney transplantation patients. Whole blood was incubated with a T-cell stimulus, after which T-cell proliferation, T-cell activation marker expression and cytokine production were measured to study residual immune activity in vitro (before drug intake; drug added to the incubation) and ex vivo (after drug intake). T-cell proliferation was completely suppressed in all patients over the full day, while IL-2, IFN-γ, CD71, and CD154 showed fluctuations over the day with a strong inhibition (75%-25%) at 2 h post-dose. The level of inhibition was variable between patients and could not be related to pharmacokinetic parameters or the presence of regulatory or senescence immune cells. Moreover, the level of inhibition did not correlate with the in vitro tacrolimus drug effect as studied by incubating pre-dose blood samples with additional tacrolimus. Overall, IL-2, IFN-γ, CD71, and CD154 seem to be good markers to monitor residual immune activity of transplantation patients. To evaluate the correlation between these pharmacodynamic biomarkers and clinical outcome, prospective observational studies are needed.


Subject(s)
Cell Proliferation , Drug Monitoring , Immunosuppressive Agents , Kidney Transplantation , Lymphocyte Activation , T-Lymphocytes , Tacrolimus , Humans , Male , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/therapeutic use , Middle Aged , Female , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Adult , Tacrolimus/administration & dosage , Tacrolimus/pharmacokinetics , Tacrolimus/pharmacology , Lymphocyte Activation/drug effects , Drug Monitoring/methods , Aged , Mycophenolic Acid/administration & dosage , Mycophenolic Acid/pharmacokinetics , Interferon-gamma/metabolism
7.
Front Immunol ; 15: 1387945, 2024.
Article in English | MEDLINE | ID: mdl-38887281

ABSTRACT

Introduction: The standard treatment for preventing rejection in vascularized composite allotransplantation (VCA) currently relies on systemic immunosuppression, which exposes the host to well-known side effects. Locally administered immunosuppression strategies have shown promising results to bypass this hurdle. Nevertheless, their progress has been slow, partially attributed to a limited understanding of the essential mechanisms underlying graft rejection. Recent discoveries highlight the crucial involvement of innate immune components, such as neutrophil extracellular traps (NETs), in organ transplantation. Here we aimed to prolong graft survival through a tacrolimus-based drug delivery system and to understand the role of NETs in VCA graft rejection. Methods: To prevent off-target toxicity and promote graft survival, we tested a locally administered tacrolimus-loaded on-demand drug delivery system (TGMS-TAC) in a multiple MHC-mismatched porcine VCA model. Off-target toxicity was assessed in tissue and blood. Graft rejection was evaluated macroscopically while the complement system, T cells, neutrophils and NETs were analyzed in graft tissues by immunofluorescence and/or western blot. Plasmatic levels of inflammatory cytokines were measured using a Luminex magnetic-bead porcine panel, and NETs were measured in plasma and tissue using DNA-MPO ELISA. Lastly, to evaluate the effect of tacrolimus on NET formation, NETs were induced in-vitro in porcine and human peripheral neutrophils following incubation with tacrolimus. Results: Repeated intra-graft administrations of TGMS-TAC minimized systemic toxicity and prolonged graft survival. Nevertheless, signs of rejection were observed at endpoint. Systemically, there were no increases in cytokine levels, complement anaphylatoxins, T-cell subpopulations, or neutrophils during rejection. Yet, tissue analysis showed local infiltration of T cells and neutrophils, together with neutrophil extracellular traps (NETs) in rejected grafts. Interestingly, intra-graft administration of tacrolimus contributed to a reduction in both T-cellular infiltration and NETs. In fact, in-vitro NETosis assessment showed a 62-84% reduction in NETs after stimulated neutrophils were treated with tacrolimus. Conclusion: Our data indicate that the proposed local delivery of immunosuppression avoids off-target toxicity while prolonging graft survival in a multiple MHC-mismatch VCA model. Furthermore, NETs are found to play a role in graft rejection and could therefore be a potential innovative therapeutic target.


Subject(s)
Drug Delivery Systems , Extracellular Traps , Graft Rejection , Graft Survival , Neutrophils , Tacrolimus , Vascularized Composite Allotransplantation , Extracellular Traps/immunology , Extracellular Traps/drug effects , Animals , Graft Survival/drug effects , Swine , Graft Rejection/immunology , Graft Rejection/prevention & control , Tacrolimus/administration & dosage , Neutrophils/immunology , Neutrophils/drug effects , Vascularized Composite Allotransplantation/methods , Immunosuppressive Agents/administration & dosage , T-Lymphocytes/immunology , Humans , Composite Tissue Allografts/immunology , Female
8.
Iran J Kidney Dis ; 18(3): 187-194, 2024 May.
Article in English | MEDLINE | ID: mdl-38904339

ABSTRACT

INTRODUCTION: Tacrolimus is the mainstem of immunosuppressive therapy in kidney transplant patients. It has high intrapatient variability (Tac-IPV), which has been reported to affect graft function by predisposing patients to rejection or nephrotoxicity. We conducted this study with the aim of assessing the influence of Tac-IPV on 2-year graft function, biopsy-proven rejection, and infections in compliant renal recipients. METHODS: In this single-center retrospective analytic cross-sectional study, 250 patients who underwent transplantation from March 21, 2018, to March 20, 2020 and had at least three outpatient tacrolimus trough levels on the same daily dose 6 to 12 months after transplantation were recruited. Tac-IPV was defined as a coefficient variation of > 15%. Graft function, biopsy-proven rejection, cytomegalovirus (CMV) and BK virus viremia, and calcineurin inhibitor (CNI) toxicity were evaluated. RESULTS: Of 202 transplant recipients, 128 were included with a mean age of 45.48 ± 13.14 years. The median Tac-IPV was 13.28% with 43.75% of patients with Tac-IPV > 15%. There were no significant differences in graft function, rejection, CNI toxicity, and CMV viremia among the groups during the 24-month study (P > .05).  However, BK viremia was significantly higher among patients with Tac-IPV > 15% (13 vs. 2.9%, P = .042). The risk of antibody mediated rejection alone (22.7 vs. 2.9%) or any kind of rejection (22.7 vs. 11.8%) was significantly higher in patients with higher Tac-IPV, and in those who had mean trough levels below 7 ng/mL (P = .015, .032; respectively). CONCLUSION: Tac-IPV is low in adherent patients (with the median of 13.28%) and maintaining tacrolimus trough level above 7 ng/mL can overcome the adverse graft outcome of Tac-IPV in compliant kidney transplant recipients. DOI: 10.52547/ijkd.7815.


Subject(s)
Graft Rejection , Immunosuppressive Agents , Kidney Transplantation , Tacrolimus , Humans , Kidney Transplantation/adverse effects , Tacrolimus/therapeutic use , Tacrolimus/adverse effects , Tacrolimus/pharmacokinetics , Female , Male , Cross-Sectional Studies , Middle Aged , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/administration & dosage , Retrospective Studies , Adult , Graft Rejection/prevention & control , Graft Rejection/immunology , Cytomegalovirus Infections , Graft Survival/drug effects , Medication Adherence , Polyomavirus Infections , Calcineurin Inhibitors/adverse effects , Calcineurin Inhibitors/administration & dosage , Calcineurin Inhibitors/therapeutic use , Calcineurin Inhibitors/pharmacokinetics , Viremia
9.
Sci Rep ; 14(1): 14214, 2024 06 20.
Article in English | MEDLINE | ID: mdl-38902302

ABSTRACT

Previous studies showed tacrolimus monotherapy and dual therapy with tacrolimus and prednisone as effective treatment modalities in managing membranous nephropathy. However, few studies have compared these therapeutic regimens. The patients were divided into two groups based on the treatment regimen: (1) tacrolimus and prednisone dual therapy (T + P group, n = 67) treatment group; and (2) tacrolimus monotherapy (T group, n = 65) or the control group. Propensity matching method and subgroup analysis to eliminate the bias in the relationship between the treatment regimen and the outcomes. The mean remission times were 20.33 ± 2.75 weeks at T group and 9.50 ± 1.81 weeks at T + P group. The T group had a remission rates of 73.33, 76.66 and 66.66% at 12weeks, 24weeks and 48weeks, while the T + P group had a remission rate of 81.66, 86.66, 91.66%; At the follow-up of 48 weeks, the relapse rate for the T group was 21.66%, and that for the T + P group was 5%. The anti-PLA2R ab is positive and therapy may be the independent risk factors for predicting remission. Tacrolimus and low-dose prednisone dual therapy is efficacious in managing MN and lowers the recurrence rate in clinical practice.


Subject(s)
Drug Therapy, Combination , Glomerulonephritis, Membranous , Immunosuppressive Agents , Prednisone , Tacrolimus , Humans , Tacrolimus/therapeutic use , Tacrolimus/administration & dosage , Glomerulonephritis, Membranous/drug therapy , Prednisone/therapeutic use , Prednisone/administration & dosage , Male , Female , Middle Aged , Retrospective Studies , Immunosuppressive Agents/therapeutic use , Adult , Treatment Outcome , Aged , Remission Induction
10.
Tissue Eng Regen Med ; 21(5): 723-735, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38834902

ABSTRACT

BACKGROUND: A drug-eluting stent (DES) is a highly beneficial medical device used to widen or unblock narrowed blood vessels. However, the drugs released by the implantation of DES may hinder the re-endothelialization process, increasing the risk of late thrombosis. We have developed a tacrolimus-eluting stent (TES) that as acts as a potent antiproliferative and immunosuppressive agent, enhancing endothelial regeneration. In addition, we assessed the safety and efficacy of TES through both in vitro and in vivo tests. METHODS: Tacrolimus and Poly(lactic-co-glycolic acid) (PLGA) were applied to the metal stent using electrospinning equipment. The surface morphology of the stent was examined before and after coating using a scanning electron microscope (SEM) and energy dispersive X-rays (EDX). The drug release test was conducted through high-performance liquid chromatography (HPLC). Cell proliferation and migration assays were performed using smooth muscle cells (SMC). The stent was then inserted into the porcine coronary artery and monitored for a duration of 4 weeks. RESULTS: SEM analysis confirmed that the coating surface was uniform. Furthermore, EDX analysis showed that the surface was coated with both polymer and drug components. The HPCL analysis of TCL at a wavelength of 215 nm revealed that the drug was continuously released over a period of 4 weeks. Smooth muscle cell migration was significantly decreased in the tacrolimus group (54.1% ± 11.90%) compared to the non-treated group (90.1% ± 4.86%). In animal experiments, the stenosis rate was significantly reduced in the TES group (29.6% ± 7.93%) compared to the bare metal stent group (41.3% ± 10.18%). Additionally, the fibrin score was found to be lower in the TES group compared to the group treated with a sirolimus-eluting stent (SES). CONCLUSION: Similar to SES, TES reduces neointimal proliferation in a porcine coronary artery model, specifically decreasing the fibrins score. Therefore, tacrolimus could be considered a promising drug for reducing restenosis and thrombosis.


Subject(s)
Cell Proliferation , Coronary Vessels , Drug-Eluting Stents , Tacrolimus , Animals , Tacrolimus/pharmacology , Coronary Vessels/drug effects , Swine , Cell Proliferation/drug effects , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/cytology , Cell Movement/drug effects
11.
Arch Dermatol Res ; 316(7): 350, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38850408

ABSTRACT

Loss and absence of melanocytes due to a number of factors is responsible for vitiligo; known to be the commonest disorder of pigmentation. The aim of the current work was to compare the efficacy and safety of excimer light with topical tacrolimus ointment 0.1% versus excimer light with topical bimatoprost gel 0.01% in treatment of facial vitiligo. The study was carried out on 48 patients presented with facial vitiligo. The patients were divided randomly using sealed envelope method into two groups (24 patients each). Group 1 were treated with excimer light plus topical tacrolimus ointment 0.1% and group 2 treated with excimer light plus topical bimatoprost gel 0.01%. Clinical improvement based on the quartile grading scale at the end of treatment did not show any statistically significant difference between groups. The majority of subjects in both groups experienced good to excellent improvement. Only 20.9% of patients in group 1 and 33.3% of subjects in group 2 achieved less than 50% repigmentation (p = 0.889). Our study demonstrated that 0.01% topical bimatoprost gel in combination with excimer light is considered safe and effective as treatment of nonsegmental facial vitiligo with comparable results to 0.1% tacrolimus.


Subject(s)
Bimatoprost , Tacrolimus , Vitiligo , Humans , Vitiligo/drug therapy , Vitiligo/therapy , Vitiligo/diagnosis , Tacrolimus/administration & dosage , Bimatoprost/administration & dosage , Female , Male , Adult , Treatment Outcome , Young Adult , Adolescent , Middle Aged , Lasers, Excimer/therapeutic use , Administration, Topical , Skin Pigmentation/drug effects , Skin Pigmentation/radiation effects , Face , Administration, Cutaneous , Child , Combined Modality Therapy/methods , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use
12.
J Clin Invest ; 134(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38828727

ABSTRACT

Calcineurin inhibitors (CNIs) constitute the backbone of modern acute graft-versus-host disease (aGVHD) prophylaxis regimens but have limited efficacy in the prevention and treatment of chronic GVHD (cGVHD). We investigated the effect of CNIs on immune tolerance after stem cell transplantation with discovery-based single-cell gene expression and T cell receptor (TCR) assays of clonal immunity in tandem with traditional protein-based approaches and preclinical modeling. While cyclosporin and tacrolimus suppressed the clonal expansion of CD8+ T cells during GVHD, alloreactive CD4+ T cell clusters were preferentially expanded. Moreover, CNIs mediated reversible dose-dependent suppression of T cell activation and all stages of donor T cell exhaustion. Critically, CNIs promoted the expansion of both polyclonal and TCR-specific alloreactive central memory CD4+ T cells (TCM) with high self-renewal capacity that mediated cGVHD following drug withdrawal. In contrast to posttransplant cyclophosphamide (PT-Cy), CSA was ineffective in eliminating IL-17A-secreting alloreactive T cell clones that play an important role in the pathogenesis of cGVHD. Collectively, we have shown that, although CNIs attenuate aGVHD, they paradoxically rescue alloantigen-specific TCM, especially within the CD4+ compartment in lymphoid and GVHD target tissues, thus predisposing patients to cGVHD. These data provide further evidence to caution against CNI-based immune suppression without concurrent approaches that eliminate alloreactive T cell clones.


Subject(s)
Calcineurin Inhibitors , Graft vs Host Disease , Isoantigens , Memory T Cells , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Graft vs Host Disease/pathology , Animals , Mice , Isoantigens/immunology , Calcineurin Inhibitors/pharmacology , Chronic Disease , Memory T Cells/immunology , Tacrolimus/pharmacology , CD4-Positive T-Lymphocytes/immunology , Cyclosporine/pharmacology , Female , CD8-Positive T-Lymphocytes/immunology , T-Lymphocyte Subsets/immunology
13.
BMC Ophthalmol ; 24(1): 251, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867175

ABSTRACT

BACKGROUND: The prevalence of rejection is 10-30% in penetrating keratoplasty (PKP) case, and the rate is higher in cases of high-risk patients. Although using topical corticosteroids is a standard method for management the rejection of post-PKP patients, it may not be sufficiently potent in high-risk patients. Topical administration of tacrolimus (TAC) may be effective in suppression rejection after corneal transplantation. This study aimed to investigate the efficacy and safety of topical TAC in high-risk PKP patients in Japan. METHODS: This study was a single centre, single-blinded, randomized controlled trial. Patients with a history of PKP, graft rejection, atopic dermatitis, or deep corneal neovascularisation who underwent PKP were enrolled. They were randomly assigned to receive 0.1% TAC ophthalmic suspension or artificial tear (AT) up to week 52 after surgery. All participants received 0.1% betamethasone up to week 13 after surgery then they received 0.1% fluorometholone up to week 52. The incidence of immunological rejection during the observation period was the main outcome measure in this study. RESULTS: Thirty patients were enrolled in this study, and 12 eyes in the TAC group and 13 eyes in the AT group completed the study, respectively. Five out of 30 patients discontinued participation after providing informed consent. No serious adverse effects were developed in patients who received 0.1% TAC ophthalmic suspension. No rejection episodes occurred in the TAC group, while one eye in the AT group had rejection. Graft clarity, best spectacle-corrected visual acuity, intraocular pressure, and corneal endothelial cell density were not significantly different between the TAC and AT groups. CONCLUSION: Our results demonstrated that good tolerability of 0.1% TAC ophthalmic suspension. However, we failed to demonstrate its efficacy in preventing immunological rejection in high-risk patients undergoing PKP. TRIAL REGISTRATION: This study was first registered in the University Hospital Medical Information Network (UMIN000029669, Date of registration: November 1, 2017). With the enforcement of the Clinical Trial Act in Japan, the study re-registered in the Japan Registry of Clinical Trials (jRCTs031180342, Date of registration: March 18, 2019).


Subject(s)
Graft Rejection , Immunosuppressive Agents , Keratoplasty, Penetrating , Ophthalmic Solutions , Tacrolimus , Humans , Tacrolimus/administration & dosage , Tacrolimus/therapeutic use , Female , Male , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Middle Aged , Graft Rejection/prevention & control , Aged , Keratoplasty, Penetrating/methods , Single-Blind Method , Administration, Topical , Visual Acuity , Adult
14.
Sci Rep ; 14(1): 12855, 2024 06 04.
Article in English | MEDLINE | ID: mdl-38834615

ABSTRACT

BK virus-associated nephropathy (BKVAN) exerts a substantial impact on allograft survival, however, the absence of robust clinical evidence regarding treatment protocols adds to the complexity of managing this condition. This study aimed to compare the two treatment approaches. The study population consisted of patients who underwent kidney transplantation between January 2016 and June 2020 at two tertiary hospitals in Korea. Patients diagnosed with BK viremia were evaluated based on their initial viral load and the treatment methods. The 'Reduction group' involved dose reduction of tacrolimus while the 'Conversion group' included tacrolimus discontinuation and conversion to sirolimus. A total of 175 patients with an initial viral load (iVL) ≥ 3 on the log10 scale were evaluated within two iVL intervals (3-4 and 4-5). In the iVL 4-5 interval, the Reduction group showed potential effectiveness in terms of viral clearance without statistically significant differences. However, within the iVL 3-4 interval, the Reduction group demonstrated superior viral clearance and a lower incidence of biopsy-proven acute rejection (BPAR) than the Conversion group. The renal function over 12 months after BKV diagnosis showed no statistically significant difference. Reducing tacrolimus compared to converting to mTORi would be a more appropriate treatment approach for BK viral clearance in kidney transplantation. Further research is warranted in a large cohort of patients.


Subject(s)
BK Virus , Calcineurin Inhibitors , Immunosuppressive Agents , Kidney Transplantation , Polyomavirus Infections , TOR Serine-Threonine Kinases , Tacrolimus , Viremia , Humans , Kidney Transplantation/adverse effects , Male , Female , Middle Aged , Calcineurin Inhibitors/therapeutic use , Viremia/drug therapy , Polyomavirus Infections/drug therapy , Tacrolimus/therapeutic use , Adult , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Immunosuppressive Agents/therapeutic use , Viral Load/drug effects , Treatment Outcome , Tumor Virus Infections/drug therapy , Tumor Virus Infections/virology , Graft Rejection/drug therapy , Graft Rejection/prevention & control , Sirolimus/therapeutic use , Republic of Korea , Retrospective Studies , Aged
15.
Int J Biol Macromol ; 273(Pt 1): 133005, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38866268

ABSTRACT

Atopic dermatitis (AD) is a chronic cutaneous disease with a complex underlying mechanism, and it cannot be completely cured. Thus, most treatment strategies for AD aim at relieving the symptoms. Although corticosteroids are topically applied to alleviate AD, adverse side effects frequently lead to the withdrawal of AD therapy. Tacrolimus (TAC), a calcineurin inhibitor, has been used to treat AD, but its high molecular weight and insolubility in water hinder its skin permeability. Herein, we developed and optimized TAC-loaded chitosan-based nanoparticles (TAC@CNPs) to improve the skin permeability of TAC by breaking the tight junctions in the skin. The prepared nanoparticles were highly loadable and efficient and exhibited appropriate characteristics for percutaneous drug delivery. TAC@CNP was stable for 4 weeks under physiological conditions. CNP released TAC in a controlled manner, with enhanced skin penetration observed. In vitro experiments showed that CNP was non-toxic to keratinocyte (HaCaT) cells, and TAC@CNP dispersed in an aqueous solution was as anti-proliferative as TAC solubilized in a good organic solvent. Importantly, an in vivo AD mouse model revealed that topical TAC@CNP containing ~1/10 of the dose of TAC found in commercially used Protopic® Ointment exhibited similar anti-inflammatory activity to that of the commercial product. TAC@CNP represents a potential therapeutic strategy for the management of AD.


Subject(s)
Chitosan , Dermatitis, Atopic , Nanoparticles , Tacrolimus , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/pathology , Tacrolimus/chemistry , Tacrolimus/pharmacology , Tacrolimus/administration & dosage , Tacrolimus/pharmacokinetics , Tacrolimus/therapeutic use , Chitosan/chemistry , Animals , Nanoparticles/chemistry , Mice , Humans , Drug Carriers/chemistry , Skin/drug effects , Skin/pathology , Skin/metabolism , Administration, Topical , Skin Absorption/drug effects , Drug Liberation , Disease Models, Animal , HaCaT Cells
16.
Biomolecules ; 14(6)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38927074

ABSTRACT

Diabetes imposes a huge burden worldwide. Islet transplantation is an alternative therapy for diabetes. However, tacrolimus, a kind of immunosuppressant after organ transplantation, is closely related to post-transplant diabetes mellitus. Mesenchymal stem cells (MSCs) have attracted interest for their potential to alleviate diabetes. In vivo experiments revealed that human menstrual blood-derived stem cells (MenSCs) treatment improved tacrolimus-induced blood glucose, body weight, and glucose tolerance disorders in mice. RNA sequencing was used to analyze the potential therapeutic targets of MenSCs. In this study, we illustrated that cystathionine ß-synthase (CBS) contributed to tacrolimus -induced islet dysfunction. Using ß-cell lines (MIN6, ß-TC-6), we demonstrated that MenSCs ameliorated tacrolimus-induced islet dysfunction in vitro. Moreover, MenSC reduced the tacrolimus-induced elevation of CBS levels and significantly enhanced the viability, anti-apoptotic ability, glucose-stimulated insulin secretion (GSIS), and glycolytic flux of ß-cells. We further revealed that MenSCs exerted their therapeutic effects by inhibiting CBS expression to activate the IL6/JAK2/STAT3 pathway. In conclusion, we showed that MenSCs may be a potential strategy to improve tacrolimus-induced islet dysfunction.


Subject(s)
Cystathionine beta-Synthase , Interleukin-6 , STAT3 Transcription Factor , Tacrolimus , Humans , STAT3 Transcription Factor/metabolism , Tacrolimus/pharmacology , Interleukin-6/metabolism , Animals , Mice , Female , Cystathionine beta-Synthase/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Janus Kinase 2/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Menstruation/blood , Menstruation/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Signal Transduction/drug effects , Insulin Secretion/drug effects , Cell Line
17.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38928269

ABSTRACT

The FUT2 gene encodes an enzyme called α-1,2-fucosyltransferase, which is involved in the formation of blood group antigens AB0(H) and is also involved in the processes of vitamin B12 absorption and its transport between cells. FUT2 gene polymorphisms are associated with vitamin B12 levels in the body. Vitamin B12 deficiency associated with hyperhomocysteinemia is a major risk factor for cardiovascular diseases (CVDs), which are one of the main causes of death in patients after kidney transplantation. The aim of our study was to determine the impact of the rs602662 (G>A) polymorphism of the FUT2 gene on the functionality of transplanted kidneys and the risk of CVD in patients after kidney transplantation. The study included 402 patients treated with immunosuppression (183 patients taking cyclosporine (CsA) and 219 patients taking tacrolimus (TAC)). The analysis of the FUT2 rs602662 (G>A) polymorphism was performed using real-time PCR. Patients with CsA were more likely to be underweight (1.64% vs. 0.91%) and obese (27.87% vs. 15.98%), while those taking TAC were more likely to be of normal weight (39.27%) or overweight (43.84%). No statistically significant differences were observed comparing the mean blood pressure, both systolic and diastolic. The renal profile showed a higher median urea nitrogen concentration in patients with CsA (26.45 mg/dL (20.60-35.40) vs. 22.95 mg/dL (17.60-33.30), p = 0.004). The observed frequency of rs602662 alleles of the FUT2 gene was similar in the analyzed groups. The A allele was present in 43.7% of patients with CsA and 41.1% of those taking TAC (OR = 0.898; 95% CI: 0.678-1.189; p = 0.453). In the group with CsA, the GG genotype was present in 32.2% of patients, the GA in 48.1% and the AA in 19.7%. A similar distribution was obtained in the TAC group: GG-33.8%, GA-50.2%, and AA-16.0%. An association of genotypes containing the G allele with a higher incidence of hypertension was observed. The G allele was present in 65% of people with hypertension and in 56% of patients with normal blood pressure (p = 0.036). Moreover, the evaluation of the renal parameters showed no effect of the FUT2 polymorphism on the risk of organ rejection because the levels of creatinine, eGFR, potassium, and urea nitrogen were prognostic of successful transplantation. Our results suggest that the rs6022662 FUT2 polymorphism may influence the risk of cardiovascular diseases.


Subject(s)
Cardiovascular Diseases , Fucosyltransferases , Galactoside 2-alpha-L-fucosyltransferase , Kidney Transplantation , Polymorphism, Single Nucleotide , Humans , Fucosyltransferases/genetics , Kidney Transplantation/adverse effects , Male , Female , Cardiovascular Diseases/genetics , Cardiovascular Diseases/etiology , Middle Aged , Adult , Risk Factors , Genetic Predisposition to Disease , Genotype , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/adverse effects , Cyclosporine/therapeutic use , Cyclosporine/adverse effects , Tacrolimus/adverse effects , Tacrolimus/therapeutic use
18.
Clin Pharmacokinet ; 63(6): 871-884, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38842789

ABSTRACT

BACKGROUND: Pharmacogenetic profiling and therapeutic drug monitoring (TDM) have both been proposed to manage inter-individual variability (IIV) in drug exposure. However, determining the most effective approach for estimating exposure for a particular drug remains a challenge. This study aimed to quantitatively assess the circumstances in which pharmacogenetic profiling may outperform TDM in estimating drug exposure, under three sources of variability (IIV, inter-occasion variability [IOV], and residual unexplained variability [RUV]). METHODS: Pharmacokinetic models were selected from the literature corresponding to drugs for which pharmacogenetic profiling and TDM are both clinically considered approaches for dose individualization. The models were used to simulate relevant drug exposures (trough concentration or area under the curve [AUC]) under varying degrees of IIV, IOV, and RUV. RESULTS: Six drug cases were selected from the literature. Model-based simulations demonstrated that the percentage of patients for whom pharmacogenetic exposure prediction is superior to TDM differs for each drug case: tacrolimus (11.0%), tamoxifen (12.7%), efavirenz (49.2%), vincristine (49.6%), risperidone (48.1%), and 5-fluorouracil (5-FU) (100%). Generally, in the presence of higher unexplained IIV in combination with lower RUV and IOV, exposure was best estimated by TDM, whereas, under lower unexplained IIV in combination with higher IOV or RUV, pharmacogenetic profiling was preferred. CONCLUSIONS: For the drugs with relatively low RUV and IOV (e.g., tamoxifen and tacrolimus), TDM estimated true exposure the best. Conversely, for drugs with similar or lower unexplained IIV (e.g., efavirenz or 5-FU, respectively) combined with relatively high RUV, pharmacogenetic profiling provided the most accurate estimate for most patients. However, genotype prevalence and the relative influence of genotypes on the PK, as well as the ability of TDM to accurately estimate AUC with a limited number of samples, had an impact. The results could be used to support clinical decision making when considering other factors, such as the probability for severe side effects.


Subject(s)
Drug Monitoring , Pharmacogenomic Testing , Humans , Drug Monitoring/methods , Pharmacogenomic Testing/methods , Tacrolimus/pharmacokinetics , Tacrolimus/therapeutic use , Tacrolimus/administration & dosage , Tamoxifen/pharmacokinetics , Tamoxifen/therapeutic use , Tamoxifen/blood , Area Under Curve , Vincristine/pharmacokinetics , Vincristine/therapeutic use , Models, Biological , Computer Simulation , Alkynes , Cyclopropanes , Benzoxazines
19.
Int Immunopharmacol ; 137: 112535, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38908078

ABSTRACT

Tacrolimus (TAC) has high pharmacokinetic (PK) variability during the early transplantation period. The relationships between whole-blood and intracellular TAC concentrations and clinical outcomes remain controversial. This study identifies the factors affecting the PK variability of TAC and characterizes the relationships between whole-blood and intracellular TAC concentrations. Data regarding whole-blood TAC concentrations of 1,787 samples from 215 renal transplant recipients (<90 days postoperative) across two centers and intracellular TAC concentrations (648 samples) digitized from previous studies were analyzed using nonlinear mixed-effects modeling. The effects of potential covariates were screened, and the distribution of whole-blood to intracellular TAC concentration ratios (RWB:IC) was estimated. The final model was evaluated using bootstrap, goodness of fit, and prediction-corrected visual predictive checks. The optimal dosing regimens and target ranges for each type of immune cell subsets were determined using Monte Carlo simulations. A two-compartment model adequately described the data, and the estimated mean TAC CL/F was 23.6 L·h-1 (relative standard error: 11.5 %). The hematocrit level, CYP3A5*3 carrier status, co-administration with Wuzhi capsules, and tapering prednisolone dose may contribute to the high variability of TAC PK variability during the early post-transplant period. The estimated RWB:IC of all TAC concentrations in peripheral blood mononuclear cells (PBMCs) was 4940, and inter-center variability of PBMCs was observed. The simulated TAC target range in PBMCs was 20.2-85.9 pg·million cells-1. Inter-center variability in intracellular concentrations should be taken into account in further analyses. TAC dosage adjustments can be guided based on PK/PD variability and simulated intracellular concentrations.


Subject(s)
Immunosuppressive Agents , Kidney Transplantation , Tacrolimus , Humans , Tacrolimus/pharmacokinetics , Tacrolimus/administration & dosage , Tacrolimus/blood , Male , Adult , Female , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/administration & dosage , Middle Aged , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A/genetics , Models, Biological , Transplant Recipients , Aged , Prednisolone/pharmacokinetics , Prednisolone/administration & dosage , Young Adult , Graft Rejection/immunology , Graft Rejection/prevention & control , Monte Carlo Method
20.
Stem Cell Res Ther ; 15(1): 167, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38872206

ABSTRACT

BACKGROUND: Stem cell therapy is a promising alternative for inflammatory diseases and tissue injury treatment. Exogenous delivery of mesenchymal stem cells is associated with instant blood-mediated inflammatory reactions, mechanical stress during administration, and replicative senescence or change in phenotype during long-term culture in vitro. In this study, we aimed to mobilize endogenous hematopoietic stem cells (HSCs) using AMD-3100 and provide local immune suppression using FK506, an immunosuppressive drug, for the treatment of inflammatory bowel diseases. METHODS: Reactive oxygen species (ROS)-responsive FK506-loaded thioketal microspheres were prepared by emulsification solvent-evaporation method. Thioketal vehicle based FK506 microspheres and AMD3100 were co-administered into male C57BL6/J mice with dextran sulfate sodium (DSS) induced colitis. The effect of FK506-loaded thioketal microspheres in colitis mice were evaluated using disease severity index, myeloperoxidase activity, histology, flow cytometry, and gene expression by qRT-PCR. RESULTS: The delivery of AMD-3100 enhanced mobilization of HSCs from the bone marrow into the inflamed colon of mice. Furthermore, targeted oral delivery of FK506 in an inflamed colon inhibited the immune activation in the colon. In the DSS-induced colitis mouse model, the combination of AMD-3100 and FK506-loaded thioketal microspheres ameliorated the disease, decreased immune cell infiltration and activation, and improved body weight, colon length, and epithelial healing process. CONCLUSION: This study shows that the significant increase in the percentage of mobilized hematopoietic stem cells in the combination therapy of AMD and oral FK506 microspheres may contribute to a synergistic therapeutic effect. Thus, low-dose local delivery of FK506 combined with AMD3100 could be a promising alternative treatment for inflammatory bowel diseases.


Subject(s)
Benzylamines , Colitis , Cyclams , Dextran Sulfate , Mice, Inbred C57BL , Tacrolimus , Animals , Colitis/chemically induced , Colitis/therapy , Colitis/drug therapy , Colitis/pathology , Mice , Male , Cyclams/pharmacology , Cyclams/therapeutic use , Tacrolimus/pharmacology , Tacrolimus/therapeutic use , Hematopoietic Stem Cell Mobilization/methods , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/therapeutic use , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Disease Models, Animal , Immunosuppression Therapy , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Microspheres , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL