Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 187
Filter
1.
Neurotherapeutics ; 20(5): 1263-1274, 2023 09.
Article in English | MEDLINE | ID: mdl-37460763

ABSTRACT

Thyroid hormones are essential during developmental myelination and may play a direct role in remyelination and repair in the adult central nervous system by promoting the differentiation of oligodendrocyte precursor cells into mature oligodendrocytes. Since tri-iodothyronine (T3) is believed to mediate the majority of important thyroid hormone actions, liothyronine (synthetic T3) has the potential to induce reparative mechanisms and limit neurodegeneration in multiple sclerosis (MS). We completed a phase 1b clinical trial to determine the safety and tolerability of ascending doses of liothyronine in individuals with relapsing and progressive MS. A total of 20 people with MS were enrolled in this single-center trial of oral liothyronine. Eighteen participants completed the 24-week study. Our study cohort included mostly women (11/20), majority relapsing MS (12/20), mean age of 46, and baseline median EDSS of 3.5. Liothyronine was tolerated well without treatment-related severe/serious adverse events or evidence of disease activation/clinical deterioration. The most common adverse events included gastrointestinal distress and abnormal thyroid function tests. No clinical thyrotoxicosis occurred. Importantly, we did not observe a negative impact on secondary clinical outcome measures. The CSF proteomic changes suggest a biological effect of T3 treatment within the CNS. We noted changes primarily in proteins associated with immune cell function and angiogenesis. Liothyronine appeared safe and was well tolerated in people with MS. A larger clinical trial will help assess whether liothyronine can promote oligodendrogenesis and enhance remyelination in vivo, limit axonal degeneration, or improve function.


Subject(s)
Multiple Sclerosis , Triiodothyronine , Female , Humans , Male , Central Nervous System , Multiple Sclerosis/drug therapy , Oligodendroglia/physiology , Proteomics , Triiodothyronine/adverse effects , Middle Aged
2.
Thyroid ; 32(8): 879-896, 2022 08.
Article in English | MEDLINE | ID: mdl-35403448

ABSTRACT

Background: Low levels of the active thyroid hormone triiodothyronine (T3) in cardiac patients are associated with worse outcomes. The aim of this analysis was to assess if T3 treatment is beneficial and safe in patients undergoing cardiac surgery or those with cardiovascular diseases in whom there is observed or expected reduction in serum T3 levels. Methods: A systematic review and meta-analysis of randomized controlled trials (RCTs) was performed as per the PRISMA guidelines. Pubmed, EMBASE, and Web of Science databases were searched for RCTs published between January 1, 1960 and March 30, 2022 that evaluated the effects of T3 therapy in patients undergoing cardiac surgery or with cardiovascular diseases. The primary outcomes were measures of cardiac function. Weighted mean difference (MD) or relative risk was calculated using a random effects model. PROSPERO registration number CRD42020211966. Results: Of the 3181 full-text articles screened, 34 studies with 2547 participants (number ranging between 13 and 223, mean ages between 0.5 and 73 years, mean percentage of women between 7% and 64%) were included. In 12 RCTs with 1093 adults undergoing cardiac surgery T3 therapy was associated with improvement in cardiac index (MD [95% confidence interval], 0.24 [0.08 to 0.40] L/min/m2, I2 = 74%). The quality of evidence was high to moderate. In 3 RCTs with 188 children undergoing cardiac surgery, 3 RCTs with 131 adult cardiac donors, 3 RCTs with 83 adult patients with heart failure, and 2 RCTs with 89 adults with acute myocardial infarction, T3 therapy did not improve cardiac index or left ventricular function; the quality of evidence ranged from high (pediatric cardiac surgery) to low (other groups). No detrimental effect of T3 therapy was observed on heart rate, risk of in-hospital atrial fibrillation, or mortality. Conclusions: Short-term T3 therapy is safe and trials in adults undergoing cardiac surgical procedures to evaluate longer term clinical endpoints are required. Current data do not support the routine use of T3 therapy in children undergoing cardiac surgery or in cardiac donors. Adequately designed trials are required to determine if T3 therapy improves cardiac function and clinical outcomes in patients with heart failure or acute myocardial infarction.


Subject(s)
Cardiac Surgical Procedures , Cardiovascular Diseases , Heart Failure , Myocardial Infarction , Adolescent , Adult , Aged , Cardiac Surgical Procedures/adverse effects , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/etiology , Child , Child, Preschool , Female , Humans , Infant , Middle Aged , Myocardial Infarction/drug therapy , Randomized Controlled Trials as Topic , Triiodothyronine/adverse effects , Young Adult
3.
J Clin Endocrinol Metab ; 107(3): e1136-e1147, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34679181

ABSTRACT

CONTEXT: Patients with mutations in thyroid hormone transporter MCT8 have developmental delay and chronic thyrotoxicosis associated with being underweight and having cardiovascular dysfunction. OBJECTIVE: Our previous trial showed improvement of key clinical and biochemical features during 1-year treatment with the T3 analogue Triac, but long-term follow-up data are needed. METHODS: In this real-life retrospective cohort study, we investigated the efficacy of Triac in MCT8-deficient patients in 33 sites. The primary endpoint was change in serum T3 concentrations from baseline to last available measurement. Secondary endpoints were changes in other thyroid parameters, anthropometric parameters, heart rate, and biochemical markers of thyroid hormone action. RESULTS: From October 15, 2014 to January 1, 2021, 67 patients (median baseline age 4.6 years; range, 0.5-66) were treated up to 6 years (median 2.2 years; range, 0.2-6.2). Mean T3 concentrations decreased from 4.58 (SD 1.11) to 1.66 (0.69) nmol/L (mean decrease 2.92 nmol/L; 95% CI, 2.61-3.23; P < 0.0001; target 1.4-2.5 nmol/L). Body-weight-for-age exceeded that of untreated historical controls (mean difference 0.72 SD; 95% CI, 0.36-1.09; P = 0.0002). Heart-rate-for-age decreased (mean difference 0.64 SD; 95% CI, 0.29-0.98; P = 0.0005). SHBG concentrations decreased from 245 (99) to 209 (92) nmol/L (mean decrease 36 nmol/L; 95% CI, 16-57; P = 0.0008). Mean creatinine concentrations increased from 32 (11) to 39 (13) µmol/L (mean increase 7 µmol/L; 95% CI, 6-9; P < 0.0001). Mean creatine kinase concentrations did not significantly change. No drug-related severe adverse events were reported. CONCLUSIONS: Key features were sustainably alleviated in patients with MCT8 deficiency across all ages, highlighting the real-life potential of Triac for MCT8 deficiency.


Subject(s)
Mental Retardation, X-Linked/drug therapy , Monocarboxylic Acid Transporters/deficiency , Muscle Hypotonia/drug therapy , Muscular Atrophy/drug therapy , Symporters/deficiency , Triiodothyronine/analogs & derivatives , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Male , Mental Retardation, X-Linked/blood , Mental Retardation, X-Linked/genetics , Middle Aged , Monocarboxylic Acid Transporters/genetics , Muscle Hypotonia/blood , Muscle Hypotonia/genetics , Muscular Atrophy/blood , Muscular Atrophy/genetics , Mutation , Retrospective Studies , Symporters/genetics , Treatment Outcome , Triiodothyronine/administration & dosage , Triiodothyronine/adverse effects , Triiodothyronine/blood , Young Adult
4.
Biol Pharm Bull ; 44(12): 1824-1831, 2021.
Article in English | MEDLINE | ID: mdl-34853265

ABSTRACT

Thyroid hormones (THs) have been suggested to play an important role in both physiological and pathological events in the central nervous system. Hypothyroidism, which is characterized by low levels of serum THs, has been associated with aggravation of ischemic neuronal injuries in stroke patients. We hypothesized that administration of T3, the main active form of THs, may attenuate the ischemic neuronal injuries. In mice, global cerebral ischemia (GCI), which is induced by transient occlusion of the bilateral common carotid artery, causes neuronal injuries by inducing neuronal death and activating inflammatory responses after reperfusion in the hippocampus. In this study, we examined the effect of T3 administration on DNA fragmentation induced by neuronal death and the activation of inflammatory cells such as astrocytes and microglia in the hippocampus following GCI. The content of nucleosomes generated by DNA fragmentation in the hippocampus was increased by GCI and further increased by T3 administration. The protein expression levels of glial fibrillary acidic protein (GFAP), an astrocytic marker, and Ionized calcium binding adaptor protein 1 (Iba1), a microglial marker, in the hippocampus were also increased by GCI and further increased by T3 administration. The levels of T3 in both the serum and hippocampus were elevated by T3 administration. Our results indicate that T3 administration aggravates GCI-reperfusion injury in mice. There may be an increased risk of aggravation of ischemic stroke by the excessive elevation of T3 levels during the drug treatment of hypothyroidism.


Subject(s)
Brain Ischemia , Hippocampus/drug effects , Reperfusion Injury , Severity of Illness Index , Triiodothyronine/adverse effects , Animals , Astrocytes , Cell Death , Cerebral Infarction , DNA Fragmentation , Disease Models, Animal , Glial Fibrillary Acidic Protein , Hypothyroidism/complications , Hypothyroidism/drug therapy , Inflammation , Male , Mice, Inbred C57BL , Microglia , Neurons , Nucleosomes , Reperfusion , Triiodothyronine/blood
5.
BMC Neurol ; 21(1): 93, 2021 Feb 27.
Article in English | MEDLINE | ID: mdl-33639866

ABSTRACT

BACKGROUND: Multiple acyl-CoA dehydrogenase deficiency (MADD) is a treatable lipid metabolism disorder that presents as myopathy and episodic metabolic crisis. The metabolic crisis is typically associated with prolonged fasting or physical stress; however, the mechanism of metabolic crisis is not yet fully understood. CASE PRESENTATION: A 28-year-old Taiwanese woman presented with dyspnoea, poor appetite, and muscle weakness after using antiobesity drugs, including metformin, triiodothyronine, and topiramate. MADD was diagnosed, and her symptoms rapidly improved after treatment with riboflavin, carnitine, and ubiquinone. To date, antiobesity drugs have not been reported to be a provoking factor in fatty acid oxidation disorder. CONCLUSIONS: The increase of ß-oxidation activity due to antiobesity drugs supports the hypothetical substrate competition model for MADD metabolic crisis. Because the drugs our patient used are commonly prescribed, we report this case to increase the vigilance and proactivity of clinicians in recognising this treatable adult-onset myopathy.


Subject(s)
Anti-Obesity Agents/adverse effects , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/complications , Muscular Diseases/genetics , Adult , Asian People , Carnitine/therapeutic use , Female , Humans , Metformin/adverse effects , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/drug therapy , Muscle Weakness/chemically induced , Riboflavin/therapeutic use , Topiramate/adverse effects , Triiodothyronine/adverse effects , Ubiquinone/analogs & derivatives , Ubiquinone/therapeutic use
6.
J Pharmacol Exp Ther ; 376(1): 74-83, 2021 01.
Article in English | MEDLINE | ID: mdl-33127750

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a severe, life-threatening form of respiratory failure characterized by pulmonary edema, inflammation, and hypoxemia due to reduced alveolar fluid clearance (AFC). Alveolar fluid clearance is required for recovery and effective gas exchange, and higher rates of AFC are associated with reduced mortality. Thyroid hormones play multiple roles in lung function, and L-3,5,3'-triiodothyronine (T3) has multiple effects on lung alveolar type II cells. T3 enhances AFC in normal adult rat lungs when administered intramuscularly and in normal or hypoxia-injured lungs when given intratracheally. The safety of a commercially available formulation of liothyronine sodium (synthetic T3) administered intratracheally was assessed in an Investigational New Drug Application-enabling toxicology study in healthy rats. Instillation of the commercial formulation of T3 without modification rapidly caused tracheal injury and often mortality. Intratracheal instillation of T3 that was reformulated and brought to a neutral pH at the maximum feasible dose of 2.73 µg T3 in 300 µl for 5 consecutive days had no clinically relevant T3-related adverse clinical, histopathologic, or clinical pathology findings. There were no unscheduled deaths that could be attributed to the reformulated T3 or control articles, no differences in the lung weights, and no macroscopic or microscopic findings considered to be related to treatment with T3. This preclinical safety study has paved the way for a phase I/II study to determine the safety and tolerability of a T3 formulation delivered into the lungs of patients with ARDS, including coronavirus disease 2019-associated ARDS, and to measure the effect on extravascular lung water in these patients. SIGNIFICANCE STATEMENT: There is growing interest in treating lung disease with thyroid hormone [triiodothyronine (T3)] in pulmonary edema and acute respiratory distress syndrome (ARDS). However, there is not any published experience on the impact of direct administration of T3 into the lung. An essential step is to determine the safety of multiple doses of T3 administered in a relevant animal species. This study enabled Food and Drug Administration approval of a phase I/II clinical trial of T3 instillation in patients with ARDS, including coronavirus disease 2019-associated ARDS (T3-ARDS ClinicalTrials.gov Identifier NCT04115514).


Subject(s)
Instillation, Drug , Lung/drug effects , Respiratory Distress Syndrome/drug therapy , Triiodothyronine/adverse effects , Animals , Drug Evaluation, Preclinical , Female , Intubation, Intratracheal/adverse effects , Intubation, Intratracheal/methods , Male , Rats , Rats, Sprague-Dawley , Triiodothyronine/administration & dosage , Triiodothyronine/therapeutic use
7.
J Cell Physiol ; 236(4): 3059-3072, 2021 04.
Article in English | MEDLINE | ID: mdl-32964425

ABSTRACT

Clinical studies have shown a correlation between thyroid disorders and cardiac diseases. High levels of triiodothyronine (T3) induce cardiac hypertrophy, a risk factor for cardiac complications and heart failure. Previous results have demonstrated that angiotensin-(1-7) is able to block T3-induced cardiac hypertrophy; however, the molecular mechanisms involved in this event have not been fully elucidated. Here, we evidenced the contribution of FOXO3 signaling to angiotensin-(1-7) effects. Angiotensin-(1-7) treatment increased nuclear FOXO3 levels and reduced p-FOXO3 levels (inactive form) in isolated cardiomyocytes. Knockdown of FOXO3 by RNA silencing abrogated the antihypertrophic effect of angiotensin-(1-7). Increased expression of antioxidant enzymes superoxide dismutase 1 (SOD1 and catalase) and lower levels of reactive oxygen species and nuclear factor-κB (NF-κB) were observed after angiotensin-(1-7) treatment in vitro. Consistent with these results, transgenic rats overexpressing angiotensin-(1-7) displayed increased nuclear FOXO3 and SOD1 levels and reduced NF-κB levels in the heart. These results provide a new molecular mechanism responsible for the antihypertrophic effect of angiotensin-(1-7), which may contribute to future therapeutic targets.


Subject(s)
Angiotensin I/pharmacology , Catalase/metabolism , Forkhead Box Protein O3/metabolism , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Peptide Fragments/pharmacology , Superoxide Dismutase-1/metabolism , Triiodothyronine/adverse effects , Up-Regulation , Animals , Antioxidants/metabolism , Down-Regulation/drug effects , Hypertrophy , Male , Models, Biological , Myocytes, Cardiac/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Rats, Sprague-Dawley , Rats, Transgenic , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/metabolism , Up-Regulation/drug effects
9.
Thyroid ; 31(2): 156-182, 2021 02.
Article in English | MEDLINE | ID: mdl-33276704

ABSTRACT

Background: Fourteen clinical trials have not shown a consistent benefit of combination therapy with levothyroxine (LT4) and liothyronine (LT3). Despite the publication of these trials, combination therapy is widely used and patients reporting benefit continue to generate patient and physician interest in this area. Recent scientific developments may provide insight into this inconsistency and guide future studies. Methods: The American Thyroid Association (ATA), British Thyroid Association (BTA), and European Thyroid Association (ETA) held a joint conference on November 3, 2019 (live-streamed between Chicago and London) to review new basic science and clinical evidence regarding combination therapy with presentations and input from 12 content experts. After the presentations, the material was synthesized and used to develop Summary Statements of the current state of knowledge. After review and revision of the material and Summary Statements, there was agreement that there was equipoise for a new clinical trial of combination therapy. Consensus Statements encapsulating the implications of the material discussed with respect to the design of future clinical trials of LT4/LT3 combination therapy were generated. Authors voted upon the Consensus Statements. Iterative changes were made in several rounds of voting and after comments from ATA/BTA/ETA members. Results: Of 34 Consensus Statements available for voting, 28 received at least 75% agreement, with 13 receiving 100% agreement. Those with 100% agreement included studies being powered to study the effect of deiodinase and thyroid hormone transporter polymorphisms on study outcomes, inclusion of patients dissatisfied with their current therapy and requiring at least 1.2 µg/kg of LT4 daily, use of twice daily LT3 or preferably a slow-release preparation if available, use of patient-reported outcomes as a primary outcome (measured by a tool with both relevant content validity and responsiveness) and patient preference as a secondary outcome, and utilization of a randomized placebo-controlled adequately powered double-blinded parallel design. The remaining statements are presented as potential additional considerations. Discussion: This article summarizes the areas discussed and presents Consensus Statements to guide development of future clinical trials of LT4/LT3 combination therapy. The results of such redesigned trials are expected to be of benefit to patients and of value to inform future thyroid hormone replacement clinical practice guidelines treatment recommendations.


Subject(s)
Hypothyroidism/drug therapy , Thyroxine/therapeutic use , Triiodothyronine/therapeutic use , Consensus , Drug Combinations , Evidence-Based Medicine , Humans , Hypothyroidism/blood , Hypothyroidism/diagnosis , Thyroxine/adverse effects , Treatment Outcome , Triiodothyronine/adverse effects
10.
Trials ; 21(1): 573, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32586399

ABSTRACT

OBJECTIVES: Tissue hypoxia is the main cause of multi-organ dysfunction in sepsis. However, effective pharmacological treatments to combat sepsis-induced tissue hypoxia are not available. Emerging experimental and clinical evidence reveals an evolutionary conserved action of thyroid hormone (TH) to adapt injured tissue to hypoxic conditions via its action on p38 MAPK, Akt signaling pathways. In addition, TH has favorable effects on the immune system and viral load in infected tissue. Non-Thyroid Illness Syndrome is common in sepsis, acute myocardial infarction and trauma and is associated with increased mortality. Thus, TH may be a novel treatment in the setting of critical illness due to viral infection in which hypoxia prevails. The present study aims to address the efficacy and safety of acute administration of triiodothyronine (T3) in critically ill COVID-19 infected patients requiring mechanical respiratory support or Extra Corporeal Membrane Oxygenation (ECMO). TRIAL DESIGN: This study is a phase II, parallel, 2-arm (1:1 ratio), multi-centre, prospective, randomized, double-blind, placebo controlled trial. PARTICIPANTS: Male and female patients aged over 18 years old who are diagnosed with pulmonary infection due to COVID-19, admitted to Intensive Care Unit and requiring mechanical ventilation or ECMO will be enrolled in this trial. Patients will be excluded in cases of pregnancy, severe systemic disease with life expectancy less than 6 months, participation in another trial of an investigational drug or device, corticosteroid and/or sympathomimetic use before initiation of treatment. All data will be collected in electronic CRF files. Participants will start to be recruited from the ICU center of "ATTIKO" University Hospital in Greece. We aim to include two more clinical sites in the trial one from Greece and one from Germany INTERVENTION AND COMPARATOR: Intervention: T3 Solution for injection 10 µg/ml. The dose administered will be 0.8g/kg i.v. bolus and will be followed by an infusion of 0.113g. kg-1.h-1 i.v. for 48 hours (therapeutic dose). After the first 48h, a maintenance dose will be administered corresponding to 50% of the therapeutic dose (0.057g. kg-1.h-1 i.v.). Drug administration will stop after successful weaning or end of follow up (maximum 30 days). Comparator: Placebo with composition and dosage identical apart from the active substance. MAIN OUTCOMES: The primary outcome assessed in the present study will be the percentage of patients successfully weaned after 30 days of follow-up. Successful weaning is defined as no requirement for ventilatory support after extubation (mechanical support) or support from ECMO for 48 hours. RANDOMISATION: An allocation sequence to one of the groups will be prepared by the Sponsor of the study. A 1:1 treatment allocation will be adopted. An electronic CRF will be used incorporating IWRS in order to assure proper randomization and unblinding in emergency cases. The representative of the sponsor will get a copy of randomization codes. The information of the randomization codes will then be locked in the database until the time at which an interim analysis or final analysis is performed. BLINDING (MASKING): Participants, caregivers, and all investigators assessing the outcomes will be blinded to group assignment. NUMBERS TO BE RANDOMISED (SAMPLE SIZE): The sample size of 60 patients (that indicates 30 subjects for each group) will have 84% power to detect the estimated difference between the two study groups. The criterion for significance (alpha) has been set at 0.05 and the test is 2-tailed. TRIAL STATUS: Protocol number T3inj-02/ThySupport, version 03, May 11, 2020. The trial is not recruiting yet. The trial will start recruitment June 18th 2020. Estimated recruitment will finish June 18th, 2021. TRIAL REGISTRATION: Triiodothyronine for the Treatment of Critically Ill Patients With COVID-19 Infection (Thy-Support), ClinicalTrials.gov Identifier: NCT04348513, date of trial registration: April 16, 2020, EudraCT Identifier: 2020-001623-13, date of trial registration: April 22, 2020 FULL PROTOCOL: The full protocol is attached as an additional file, accessible from the Trials website (Additional file 1). In the interest in expediting dissemination of this material, the familiar formatting has been eliminated; this Letter serves as a summary of the key elements of the full protocol.


Subject(s)
Betacoronavirus , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Randomized Controlled Trials as Topic , Triiodothyronine/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , COVID-19 , Critical Illness , Double-Blind Method , Extracorporeal Membrane Oxygenation , Female , Humans , Male , Middle Aged , Pandemics , Prospective Studies , Respiration, Artificial , SARS-CoV-2 , Triiodothyronine/adverse effects , Young Adult , COVID-19 Drug Treatment
11.
Mult Scler Relat Disord ; 41: 102015, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32120028

ABSTRACT

BACKGROUND: Thyroid hormone promotes remyelination in multiple sclerosis (MS) animal models through a variety of mechanisms. Liothyronine (L-T3) is a short-acting thyroid hormone with demonstrated safety and tolerability for short-term and chronic use in euthyroid adults with other health conditions, but has not been studied in people with MS. The objectives of this single-center, phase I, placebo-controlled, clinical trial were to determine the safety, tolerability, and optimal dosing of L-T3 in people with MS in preparation for a phase 2 remyelination clinical trial. Secondary goals included exploration of the reliability of functional and clinical measurements of myelination in the anterior visual pathway over one week. METHODS: Groups of six clinically stable people with MS were randomized in a 4:2 ratio to receive L-T3 or placebo. The first group received 50 mcg total daily dose (TDD) of L-T3, with escalating doses of L-T3 in subsequent groups, up to potentially 150 mcg TDD in the final group. Prior to enrollment for the next dose-escalated group, all safety measures for the prior dose were reviewed. The maximum tolerated dose (MTD) was considered to be the dose below which two or more participants experienced dose limiting symptoms or one participant experienced a serious adverse event. After the MTD was reached, no further patients were enrolled. Visual evoked potentials (VEP) P100 latency with two different check sizes (17' and 34') and Sloan low contrast letter acuity (LCLA) were measured pre- and post-treatment. To determine whether there was a treatment effect, the placebo and L-T3 groups were compared using a clustered bootstrap regression estimation. A linear mixed effects model was used to determine test-retest reliability of VEP and LCLA in all eyes. RESULTS: Between May 2016 and November 2016, 15 people with MS were randomized to L-T3 (n = 10) or placebo (n = 5). Subjects were adherent to the study drug and the MTD was 75 mcg TDD. No serious adverse events were observed and the most common adverse events were poor sleep and loose stools. No treatment effect of L-T3 was observed over one week. Therefore, data from patients on L-T3 and placebo were pooled to explore VEP and LCLA reliability. The intraclass correlations of VEP 17', VEP 34' and LCLA were 0.836, 0.860, and 0.932, respectively. The mean differences in values between visits 1 and 2 for VEP 17' and 34' and LCLA were 1.9 ms/eye (SD 6.5), 0.4 ms/eye (6.3), and 0.8/eye (3.6), respectively. CONCLUSIONS: This study confirms the short-term safety and tolerability of L-T3 in people with MS, with 75 mcg TDD as the MTD. Our results also support that, despite small variations over one week, VEP with various check sizes and Sloan LCLA are reliable functional and clinical outcome measures that could be used in remyelination clinical trials in MS. A future phase 2 clinical trial to investigate the efficacy of L-T3 as a remyelination therapy may be warranted. This trial was registered on clinicaltrials.gov (NCT02760056).


Subject(s)
Evoked Potentials, Visual/drug effects , Multiple Sclerosis/drug therapy , Outcome Assessment, Health Care/standards , Remyelination/drug effects , Triiodothyronine/pharmacology , Visual Acuity/drug effects , Adult , Female , Humans , Male , Middle Aged , Reproducibility of Results , Triiodothyronine/administration & dosage , Triiodothyronine/adverse effects
12.
BMJ Case Rep ; 13(1)2020 Jan 13.
Article in English | MEDLINE | ID: mdl-31937628

ABSTRACT

A 25-year-old man presented generally unwell to the emergency department. Initial assessment identified systemic inflammatory response syndrome markers with an insect bite as a potential source of infection and he was treated for presumed sepsis. Tachycardia persisted and baseline thyroid function testing showed undetectable free thyroxine and thyroid-stimulating hormone (TSH), prompting further endocrine investigation. Triiodothyronine (T3) was markedly raised with normal TSH receptor antibodies, and the patient later confessed to supplementary testosterone and T3 use as part of bodybuilding activities. Following counselling, thyroid function normalised and the patient returned to his usual health. This case describes the diagnostic work up in a case of persistent tachycardia caused by T3 supplementation, demonstrating the potential for endocrine supplementation by bodybuilders which may be poorly understood and recognised by clinicians. T3 supplementation should be considered and a thorough drug history obtained in bodybuilders presenting with symptoms of thyrotoxicosis and deranged thyroid function tests.


Subject(s)
Sepsis/chemically induced , Tachycardia/chemically induced , Thyrotoxicosis/chemically induced , Triiodothyronine/adverse effects , Adult , Athletes , Delayed Diagnosis , Diagnosis, Differential , Humans , Male , Testosterone Congeners , Thyroid Function Tests , Weight Lifting
13.
Gen Comp Endocrinol ; 289: 113382, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31917150

ABSTRACT

Seasonally spawning fish rely on a dynamic and complex hormonal interplay to regulate cycles of gonadal development and the regression. Thyroid hormones have been shown to be a key player during gonadal development, and can regulate the activity of a number of essential reproductive hormones. Apoptosis is a vital cellular process that contributes to the hormonal control of gonadal development and regression, but the roles of thyroid hormones on gonadal apoptosis in goldfish have not been explored. The present study examines the role of acute T3 exposure on caspase 3-dependent apoptosis in dispersed goldfish gonadal tissue in vitro. We examined the levels of caspase 3 activity in early, mid, and late recrudescent gonadal tissue after exposure to physiological doses of T3 for up to 24 h. Acute treatment with T3 did not alter basal caspase 3 activity in goldfish gonads in vitro in these reproductive stages. This initial study suggests that transient increases in T3 levels are unlikely to directly contribute to basal caspase 3-dependent apoptosis in the gonadal tissue of goldfish, although we cannot rule out an interaction of T3 with other hormones involved in the control of apoptosis in the testis and ovary.


Subject(s)
Caspase 3/drug effects , Goldfish/physiology , Gonads/drug effects , Triiodothyronine/adverse effects , Animals , Female , Male
14.
Semin Thorac Cardiovasc Surg ; 32(1): 87-95, 2020.
Article in English | MEDLINE | ID: mdl-31128253

ABSTRACT

Thyroid hormone modifies metabolic, immune and cardiovascular functions and has been administered perioperatively to treat a relative reduction of thyroid function in children following cardiopulmonary bypass (CPB) for correction of congenital heart disease. However, it remains unclear whether its use is associated with improved outcomes. We performed a meta-analysis of studies that evaluated the impact of thyroid hormone supplementation on clinical outcomes in children undergoing repair of congenital heart disease using CPB. A systematic review of published trials was conducted to identify studies of children randomized to thyroid hormone supplementation or placebo undergoing congenital heart surgery. A meta-analysis was then conducted to determine the clinical impact of thyroid hormone replacement on cardiac function and postoperative characteristics. The following outcomes were included for the study: duration of mechanical ventilation, duration of intensive care unit (ICU) stay, duration of postoperative hospital stay, inotrope score, cardiac index at 24 hours postoperatively, and inpatient mortality. A total of 9 studies with 711 patients were included in the analyses. All included studies were prospective and patients were randomized to either thyroid hormone or placebo. There was wide variation in thyroid hormone dosing, ranging from 0.4 µg/kg up to 5 µg/kg over a 24-hour period, and duration of therapy, ranging from a single dose after cessation of CPB to continued thyroid hormone for the duration of the ICU stay. There was a significant difference in the mean inotrope score between the 2 groups of -1.249 (95% confidence interval -1.570 to -0.929, P < 0.001), with the inotrope score being significantly lower in the thyroid group. There was no difference in duration of mechanical ventilation, duration of ICU stay, duration of hospital stay, cardiac index, and mortality between groups. In this meta-analysis, routine thyroid hormone replacement with approximately 1-5 µg/kg administered over 24 hours does not significantly alter the postoperative course in children following CPB. However, given a clinically small but significant difference in respect to lower inotrope score and shorter duration of ICU and hospital stays with higher thyroid replacement additional studies are warranted.


Subject(s)
Cardiac Surgical Procedures , Cardiopulmonary Bypass , Heart Defects, Congenital/surgery , Hormone Replacement Therapy , Hypothyroidism/drug therapy , Triiodothyronine/administration & dosage , Age Factors , Cardiac Surgical Procedures/adverse effects , Cardiac Surgical Procedures/mortality , Cardiopulmonary Bypass/adverse effects , Cardiopulmonary Bypass/mortality , Female , Heart Defects, Congenital/mortality , Hormone Replacement Therapy/adverse effects , Hormone Replacement Therapy/mortality , Hospital Mortality , Humans , Hypothyroidism/diagnosis , Hypothyroidism/etiology , Hypothyroidism/mortality , Infant , Infant, Newborn , Length of Stay , Male , Postoperative Care , Randomized Controlled Trials as Topic , Recovery of Function , Risk Factors , Time Factors , Treatment Outcome , Triiodothyronine/adverse effects
15.
Mol Cell Endocrinol ; 493: 110451, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31112742

ABSTRACT

Ubiquitin proteasome system (UPS) is the main proteolytic pathway in eukaryotic cells. Changes in proteasome expression and activity have been associated to cardiovascular diseases as cardiac hypertrophy. Considering that cardiac hypertrophy is commonly associated to hyperthyroidism condition, the present study aimed to investigate the contribution of UPS in cardiac hypertrophy induced by thyroid hormones. Hyperthyroidism was induced in male Wistar rats by intraperitoneal injections of triiodothyronine (T3; 7  µg/100 g of body weight) for 7 days and confirmed by raised levels of total T3 and decreased levels of total T4. In addition, systolic blood pressure and heart rate were significantly increased in hyperthyroid group. Cardiac hypertrophy was confirmed in hyperthyroid group by increased heart weight/tibia length ratio and by increased α-MHC/ß-MHC relative expression. Both catalytic (20SPT) and regulatory subunits (19SPT) of the constitutive proteasome were upregulated in hyperthyroid hearts. In addition, the transcripts that encode immunoproteasome subunits were also elevated. Furthermore, ATP-dependent chymotrypsin-like activity (26SPT) was significantly increased in hyperthyroid group. Despite the upregulation and activation of UPS in hyperthyroid hearts, the content of polyubiquitinated proteins was unaltered in relation to control. Together, these results evidence the activation of cardiac proteasome by thyroid hormones, which possibly contribute to the maintenance of protein quality control and regulation of cardiac hypertrophy in response to thyroid hormones.


Subject(s)
Cardiomegaly/genetics , Hyperthyroidism/genetics , Proteasome Endopeptidase Complex/genetics , Up-Regulation , Animals , Cardiomegaly/chemically induced , Disease Models, Animal , Gene Expression Regulation/drug effects , Hyperthyroidism/chemically induced , Male , Myosin Heavy Chains/genetics , Rats , Rats, Wistar , Triiodothyronine/adverse effects , Ubiquitination
16.
Regul Toxicol Pharmacol ; 106: 111-136, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31018155

ABSTRACT

Thyroid hormones (THs; T3 and T4) play a role in development of cardiovascular, reproductive, immune and nervous systems. Thus, interpretation of TH changes from rodent studies (during pregnancy, in fetuses, neonates, and adults) is critical in hazard characterization and risk assessment. A roundtable session at the 2017 Society of Toxicology (SOT) meeting brought together academic, industry and government scientists to share knowledge and different perspectives on technical and data interpretation issues. Data from a limited group of laboratories were compiled for technical discussions on TH measurements, including good practices for reliable serum TH data. Inter-laboratory historical control data, derived from immunoassays or mass spectrometry methods, revealed: 1) assay sensitivities vary within and across methodologies; 2) TH variability is similar across animal ages; 3) laboratories generally achieve sufficiently sensitive TH quantitation levels, although issues remain for lower levels of serum TH and TSH in fetuses and postnatal day 4 pups; thus, assay sensitivity is critical at these life stages. Best practices require detailed validation of rat serum TH measurements across ages to establish assay sensitivity and precision, and identify potential matrix effects. Finally, issues related to data interpretation for biological understanding and risk assessment were discussed, but their resolution remains elusive.


Subject(s)
Thyroid Gland/drug effects , Thyroxine/adverse effects , Triiodothyronine/adverse effects , Animals , Humans , Immunoassay , Mass Spectrometry , Risk Assessment , Thyroxine/administration & dosage , Triiodothyronine/administration & dosage
17.
Article in English | MEDLINE | ID: mdl-30553402

ABSTRACT

Supplementation with omega-3 fatty acids or thyroid hormone (T3) exhibit negative effects on inflammatory reactions in experimental animals. The aim of this work was to assess the hypothesis that docosahexaenoic acid (DHA) plus T3 co-administration enhances liver resolvin (Rv) levels as inflammation resolution mediators. Combined DHA (daily doses of 300 mg/kg for 3 consecutive days)-T3 (0.05 mg/kg at the fourth day) administration significantly increased the content of hepatic RvD1 and RvD2, without changes in that of RvE1 and RvE2, an effect that exhibits synergy when compared to the separate DHA and T3 treatments. Under these conditions, liver DHA levels increased by DHA administration were diminished when combined with T3 (p < 0.05), suggesting enhancement in resolvin D biosynthesis in extrahepatic tissues. It is concluded that co-administration of DHA and T3 rises the capacity of the liver for inflammation resolution by augmenting RvD1(2) availability, which represents an important protocol in hepatoprotection in the clinical setting.


Subject(s)
Docosahexaenoic Acids/pharmacology , Liver/drug effects , Liver/metabolism , Protective Agents/pharmacology , Triiodothyronine/pharmacology , Animals , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/metabolism , Drug Synergism , Drug Therapy, Combination , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/metabolism , Hepatitis, Animal/drug therapy , Male , Protective Agents/administration & dosage , Rats , Rats, Sprague-Dawley , Triiodothyronine/administration & dosage , Triiodothyronine/adverse effects
18.
BMJ Case Rep ; 11(1)2018 Dec 07.
Article in English | MEDLINE | ID: mdl-30567198

ABSTRACT

The thyroid hormone triiodothyronine (T3) can be used as an augmentation therapy for depression. This case involves a patient who had been initiated on liothyronine (L-T3) for this purpose and subsequently became pregnant. The exogenous T3 affected maternal thyroid hormone production, including suppression of her thyroxine (T4) levels. In pregnancy, maternal T4 is important for fetal neurodevelopment. Accordingly, use of exogenous T3 for adjunct treatment of depression in pregnant patients or those planning pregnancy requires careful laboratory monitoring of thyroid function tests. In this case, L-T3 was discontinued and the patient was eventually started on levothyroxine (L-T4) and went on to have an uneventful pregnancy.


Subject(s)
Depression/drug therapy , Pregnancy Complications/drug therapy , Selective Serotonin Reuptake Inhibitors/therapeutic use , Thyroxine/therapeutic use , Triiodothyronine/therapeutic use , Adult , Chemotherapy, Adjuvant , Depression/blood , Female , Humans , Pregnancy , Pregnancy Complications/blood , Thyroxine/blood , Triiodothyronine/adverse effects
19.
BMJ Case Rep ; 11(1)2018 Dec 13.
Article in English | MEDLINE | ID: mdl-30567254

ABSTRACT

The consumption of daily nutritional supplements has risen dramatically all over the world. Many people believe that dietary supplements, if not useful, are at least safe to fulfil small dietary gaps. Many nutritional supplements have not been approved by Federal Drug Administration due to their unregulated active ingredients, but they are available as over the counter. One of the active ingredients, exogenous triiodothyronine (T3), has been reported in dietary supplements. We present a case of sudden onset of tetraparesis. Laboratory workup showed hypokalaemia, low thyroid-stimulating hormone and thyroxine (T4) but normal T3 and thyroglobulin levels. The radioiodine uptake scan also showed reduced uptake. After aggressive serum potassium correction and stopping supplements, his condition got improved. So the suspicion of exogenous T3-induced thyrotoxic periodic paralysis was confirmed.


Subject(s)
Dietary Supplements/adverse effects , Paresis/chemically induced , Triiodothyronine/adverse effects , Adult , Humans , Hypokalemia/blood , Hypokalemia/chemically induced , Male , Paresis/blood , Thyrotropin/blood , Thyroxine/blood
20.
Bipolar Disord ; 20(7): 594-603, 2018 11.
Article in English | MEDLINE | ID: mdl-29869405

ABSTRACT

OBJECTIVES: This report describes the first comparative double-blind, placebo-controlled trial of levothyroxine (L-T4 ) and triiodothyronine (T3 ) as adjunctive treatments in rapid cycling bipolar disorder. METHODS: Thirty-two treatment-resistant, rapid cycling patients who had failed a trial of lithium were randomized into three treatment arms: L-T4 , T3 , or placebo. They were followed for ≥4 months with weekly clinical and endocrine assessments. RESULTS: There were no statistically significant differences between the groups in age, gender, duration of illness, or thyroid status. Markov chain analyses were employed to assess treatment effects on cycling patterns among mood states (euthymia, depression, mania, and mixed). Within groups, post-treatment the L-T4 group spent significantly less time depressed or in a mixed state and greater time euthymic. The T3 and placebo groups did not differ significantly pre- and post-treatment in any mood state, although the pattern of effects was the same for the T3 group as for the L-T4 group. Between groups, the L-T4 group had a significantly greater increase in time euthymic and decrease in time in the mixed state than the placebo group. Other group differences were not significant, although they were in the expected direction. CONCLUSIONS: The findings in this first double-blind study directly comparing the effects of L-T4 and T3 therapy against placebo provide evidence for the benefit of adjunctive L-T4 in alleviating resistant depression, reducing time in mixed states and increasing time euthymic. Adjunctive T3 did not show statistically significant evidence of benefit over placebo in reducing the time spent in disturbed mood states.


Subject(s)
Affect/drug effects , Bipolar Disorder , Thyroxine , Triiodothyronine , Adult , Bipolar Disorder/diagnosis , Bipolar Disorder/drug therapy , Bipolar Disorder/metabolism , Bipolar Disorder/psychology , Double-Blind Method , Female , Humans , Male , Middle Aged , Psychiatric Status Rating Scales , Thyroid Function Tests/methods , Thyroid Hormones/administration & dosage , Thyroid Hormones/adverse effects , Thyroid Hormones/metabolism , Thyroxine/administration & dosage , Thyroxine/adverse effects , Thyroxine/metabolism , Time Factors , Treatment Outcome , Triiodothyronine/administration & dosage , Triiodothyronine/adverse effects , Triiodothyronine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...