Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 300
Filter
1.
J Biol Chem ; 298(2): 101554, 2022 02.
Article in English | MEDLINE | ID: mdl-34973337

ABSTRACT

The mitochondrial pyruvate carrier (MPC) is an inner mitochondrial membrane complex that plays a critical role in intermediary metabolism. Inhibition of the MPC, especially in liver, may have efficacy for treating type 2 diabetes mellitus. Herein, we examined the antidiabetic effects of zaprinast and 7ACC2, small molecules which have been reported to act as MPC inhibitors. Both compounds activated a bioluminescence resonance energy transfer-based MPC reporter assay (reporter sensitive to pyruvate) and potently inhibited pyruvate-mediated respiration in isolated mitochondria. Furthermore, zaprinast and 7ACC2 acutely improved glucose tolerance in diet-induced obese mice in vivo. Although some findings were suggestive of improved insulin sensitivity, hyperinsulinemic-euglycemic clamp studies did not detect enhanced insulin action in response to 7ACC2 treatment. Rather, our data suggest acute glucose-lowering effects of MPC inhibition may be due to suppressed hepatic gluconeogenesis. Finally, we used reporter sensitive to pyruvate to screen a chemical library of drugs and identified 35 potentially novel MPC modulators. Using available evidence, we generated a pharmacophore model to prioritize which hits to pursue. Our analysis revealed carsalam and six quinolone antibiotics, as well as 7ACC1, share a common pharmacophore with 7ACC2. We validated that these compounds are novel inhibitors of the MPC and suppress hepatocyte glucose production and demonstrated that one quinolone (nalidixic acid) improved glucose tolerance in obese mice. In conclusion, these data demonstrate the feasibility of therapeutic targeting of the MPC for treating diabetes and provide scaffolds that can be used to develop potent and novel classes of MPC inhibitors.


Subject(s)
Anion Transport Proteins , Mitochondrial Membrane Transport Proteins , Monocarboxylic Acid Transporters , Obesity , Quinolones , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet , Glucose/metabolism , Mice , Mice, Obese , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Obesity/drug therapy , Obesity/metabolism , Pyruvic Acid/metabolism , Quinolones/pharmacology
2.
J Med Chem ; 64(4): 2046-2063, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33534563

ABSTRACT

Herein, we report the synthesis and evaluation of novel analogues of UK-5099 both in vitro and in vivo for the development of mitochondrial pyruvate carrier (MPC) inhibitors to treat hair loss. A comprehensive understanding of the structure-activity relationship was obtained by varying four positions of the hit compound, namely, the alkyl group on the N1 position, substituents on the indole core, various aromatic and heteroaromatic core structures, and various Michael acceptors. The major discovery was that the inhibitors with a 3,5-bis(trifluoromethyl)benzyl group at the N1 position were shown to have much better activity than JXL001 (UK-5099) to increase cellular lactate production. Additionally, analogue JXL069, possessing a 7-azaindole heterocycle, was also shown to have significant MPC inhibition activity, which further increases the chemical space for drug design. Finally, more than 10 analogues were tested on shaved mice by topical treatment and promoted obvious hair growth on mice.


Subject(s)
Acrylates/therapeutic use , Alopecia/drug therapy , Indoles/therapeutic use , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Monocarboxylic Acid Transporters/antagonists & inhibitors , Acrylates/chemical synthesis , Animals , Indoles/chemical synthesis , Lactic Acid/metabolism , Mice, Inbred C57BL , Molecular Structure , Structure-Activity Relationship
3.
Cell Metab ; 33(3): 629-648.e10, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33333007

ABSTRACT

The metabolic rewiring of cardiomyocytes is a widely accepted hallmark of heart failure (HF). These metabolic changes include a decrease in mitochondrial pyruvate oxidation and an increased export of lactate. We identify the mitochondrial pyruvate carrier (MPC) and the cellular lactate exporter monocarboxylate transporter 4 (MCT4) as pivotal nodes in this metabolic axis. We observed that cardiac assist device-induced myocardial recovery in chronic HF patients was coincident with increased myocardial expression of the MPC. Moreover, the genetic ablation of the MPC in cultured cardiomyocytes and in adult murine hearts was sufficient to induce hypertrophy and HF. Conversely, MPC overexpression attenuated drug-induced hypertrophy in a cell-autonomous manner. We also introduced a novel, highly potent MCT4 inhibitor that mitigated hypertrophy in cultured cardiomyocytes and in mice. Together, we find that alteration of the pyruvate-lactate axis is a fundamental and early feature of cardiac hypertrophy and failure.


Subject(s)
Anion Transport Proteins/metabolism , Cardiomegaly/pathology , Heart Failure/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Animals , Anion Transport Proteins/antagonists & inhibitors , Anion Transport Proteins/genetics , Cardiomegaly/chemically induced , Cardiomegaly/complications , Heart Failure/etiology , Heart-Assist Devices , Humans , Lactic Acid/metabolism , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/genetics , Muscle Proteins/antagonists & inhibitors , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Pyruvic Acid/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Ventricular Function, Left/physiology
4.
J Biol Chem ; 295(50): 17009-17026, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33028634

ABSTRACT

Stop codon read-through (SCR) is a process of continuation of translation beyond a stop codon. This phenomenon, which occurs only in certain mRNAs under specific conditions, leads to a longer isoform with properties different from that of the canonical isoform. MTCH2, which encodes a mitochondrial protein that regulates mitochondrial metabolism, was selected as a potential read-through candidate based on evolutionary conservation observed in the proximal region of its 3' UTR. Here, we demonstrate translational read-through across two evolutionarily conserved, in-frame stop codons of MTCH2 using luminescence- and fluorescence-based assays, and by analyzing ribosome-profiling and mass spectrometry (MS) data. This phenomenon generates two isoforms, MTCH2x and MTCH2xx (single- and double-SCR products, respectively), in addition to the canonical isoform MTCH2, from the same mRNA. Our experiments revealed that a cis-acting 12-nucleotide sequence in the proximal 3' UTR of MTCH2 is the necessary signal for SCR. Functional characterization showed that MTCH2 and MTCH2x were localized to mitochondria with a long t1/2 (>36 h). However, MTCH2xx was found predominantly in the cytoplasm. This mislocalization and its unique C terminus led to increased degradation, as shown by greatly reduced t1/2 (<1 h). MTCH2 read-through-deficient cells, generated using CRISPR-Cas9, showed increased MTCH2 expression and, consistent with this, decreased mitochondrial membrane potential. Thus, double-SCR of MTCH2 regulates its own expression levels contributing toward the maintenance of normal mitochondrial membrane potential.


Subject(s)
3' Untranslated Regions/genetics , Codon, Terminator/genetics , Membrane Potential, Mitochondrial , Mitochondrial Membrane Transport Proteins/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , Animals , Aorta/cytology , Aorta/metabolism , Base Sequence , CRISPR-Cas Systems/genetics , Cattle , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , HEK293 Cells , Humans , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Protein Isoforms , Ribosomes/metabolism
5.
Nat Commun ; 11(1): 3347, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32620768

ABSTRACT

A sharp increase in mitochondrial Ca2+ marks the activation of brown adipose tissue (BAT) thermogenesis, yet the mechanisms preventing Ca2+ deleterious effects are poorly understood. Here, we show that adrenergic stimulation of BAT activates a PKA-dependent mitochondrial Ca2+ extrusion via the mitochondrial Na+/Ca2+ exchanger, NCLX. Adrenergic stimulation of NCLX-null brown adipocytes (BA) induces a profound mitochondrial Ca2+ overload and impaired uncoupled respiration. Core body temperature, PET imaging of glucose uptake and VO2 measurements confirm a thermogenic defect in NCLX-null mice. We show that Ca2+ overload induced by adrenergic stimulation of NCLX-null BAT, triggers the mitochondrial permeability transition pore (mPTP) opening, leading to a remarkable mitochondrial swelling and cell death. Treatment with mPTP inhibitors rescue mitochondrial function and thermogenesis in NCLX-null BAT, while calcium overload persists. Our findings identify a key pathway through which BA evade apoptosis during adrenergic stimulation of uncoupling. NCLX deletion transforms the adrenergic pathway responsible for thermogenesis activation into a death pathway.


Subject(s)
Adipocytes, Brown/pathology , Adipose Tissue, Brown/metabolism , Norepinephrine/metabolism , Sodium-Calcium Exchanger/metabolism , Thermogenesis/physiology , Adipocytes, Brown/cytology , Adipocytes, Brown/drug effects , Adipose Tissue, Brown/cytology , Adrenergic Agents/pharmacology , Animals , Apoptosis/drug effects , Calcium/metabolism , Cells, Cultured , Cold Temperature/adverse effects , Cyclosporine/pharmacology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Female , Intravital Microscopy , Male , Mice , Mice, Knockout , Microscopy, Fluorescence , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Primary Cell Culture , Signal Transduction , Sodium-Calcium Exchanger/genetics , Thermogenesis/drug effects
6.
ACS Chem Biol ; 15(8): 2195-2204, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32584541

ABSTRACT

The Wnt signaling pathway regulates diverse cellular processes. ß-Catenin is one of the major components of this pathway, in which it plays a main role. Although it has been established that ß-catenin is mutated in a wide variety of tumors, there are currently no effective therapeutic agents that target ß-catenin. In this study, we searched for the compound that targets mutant ß-catenin and found DS37262926 (miclxin). Miclxin exhibited ß-catenin-dependent apoptosis in ß-catenin-mutated HCT116 cells and isogenic HCT116 (CTNNB1 Δ45/-) cells; however, this effect was not observed in isogenic HCT116 (CTNNB1 +/-) cells. Using miclxin-immobilized beads, MIC60, one of the major components of the mitochondrial contact site and cristae organizing system (MICOS) complex, was identified as a target protein of miclxin. We revealed that MIC60 dysfunction caused by miclxin induced a mitochondrial stress response in a mutant ß-catenin-dependent manner. Activation of the mitochondrial stress response was responsible for the downregulation of Bcl-2, leading to severe loss of mitochondrial membrane potential and subsequent apoptosis-inducing factor-dependent apoptosis. Our findings suggest that targeting MIC60 is a potential strategy with which tumor cells can be killed through induction of severe mitochondrial damage in a mutant ß-catenin-dependent manner.


Subject(s)
Apoptosis , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Stress, Physiological , beta Catenin/metabolism , HCT116 Cells , Humans , Wnt Signaling Pathway
7.
Biochem Pharmacol ; 177: 113995, 2020 07.
Article in English | MEDLINE | ID: mdl-32339494

ABSTRACT

Few discoveries have influenced drug discovery programs more than the finding that mitochondrial membranes undergo swings in permeability in response to cellular perturbations. The conductor of these permeability changes is the aptly named mitochondrial permeability transition pore which, although not yet precisely defined, is comprised of several integral proteins that differentially act to regulate the flux of ions, proteins and metabolic byproducts during the course of cellular physiological functions but also pathophysiological insults. Pursuit of the pore's exact identity remains a topic of keen interest, but decades of research have unearthed provocative functions for the integral proteins leading to their evaluation to develop novel therapeutics for a wide range of clinical indications. Chief amongst these targeted, integral proteins have been the Voltage Dependent Anion Channel (VDAC) and the F1FO ATP synthase. Research associated with the roles and ligands of VDAC has been extensive and we will expand upon 3 examples of ligand:VDAC interactions for consideration of drug discovery projects: Tubulin:VDAC1, Hexokinase I/II:VDAC1 and olesoxime:VDAC1. The discoveries that cyclosporine blocks mitochondrial permeability transition via binding to cyclophilin D, and that cyclophilin D is an important component of F1FO ATP synthase, has heightened interest in the F1FO ATP synthase as a focal point for drug discovery, and we will discuss 2 plausible campaigns associated with disease indications. To date no drug has emerged from prospective targeting these integral proteins; however, continued exploration such as the approaches suggested in this Commentary will increase the likelihood of providing important therapeutics for severely unmet medical needs.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Cholestenones/therapeutic use , Cyclosporine/therapeutic use , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/genetics , Voltage-Dependent Anion Channel 1/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Cyclophilins/genetics , Cyclophilins/metabolism , Gene Expression Regulation , Hexokinase/genetics , Hexokinase/metabolism , Humans , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Permeability/drug effects , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tubulin/genetics , Tubulin/metabolism , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/metabolism
8.
Oncogene ; 39(1): 164-175, 2020 01.
Article in English | MEDLINE | ID: mdl-31462712

ABSTRACT

Citrin, encoded by SLC25A13 gene, is an inner mitochondrial transporter that is part of the malate-aspartate shuttle, which regulates the NAD+/NADH ratio between the cytosol and mitochondria. Citrullinemia type II (CTLN-II) is an inherited disorder caused by germline mutations in SLC25A13, manifesting clinically in growth failure that can be alleviated by dietary restriction of carbohydrates. The association of citrin with glycolysis and NAD+/NADH ratio led us to hypothesize that it may play a role in carcinogenesis. Indeed, we find that citrin is upregulated in multiple cancer types and is essential for supplementing NAD+ for glycolysis and NADH for oxidative phosphorylation. Consequently, citrin deficiency associates with autophagy, whereas its overexpression in cancer cells increases energy production and cancer invasion. Furthermore, based on the human deleterious mutations in citrin, we found a potential inhibitor of citrin that restricts cancerous phenotypes in cells. Collectively, our findings suggest that targeting citrin may be of benefit for cancer therapy.


Subject(s)
Carcinogenesis/genetics , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/genetics , Neoplasms/genetics , Carbohydrates/genetics , Citrullinemia/genetics , Citrullinemia/metabolism , Cytosol/metabolism , Cytosol/pathology , Gene Expression Regulation, Neoplastic/genetics , Germ-Line Mutation/genetics , Glutamates/pharmacology , Glutamic Acid/analogs & derivatives , Glutamic Acid/pharmacology , Glycolysis/genetics , Humans , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , Oxidative Phosphorylation/drug effects
9.
BMB Rep ; 53(1): 47-55, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31818365

ABSTRACT

Alzheimer's disease (AD) is a multifactorial neurodegenerative disease and has become a major socioeconomic issue in many developed countries. Currently available therapeutic agents for AD provide only symptomatic treatments, mainly because the complete mechanism of the AD pathogenesis is still unclear. Although several different hypotheses have been proposed, mitochondrial dysfunction has gathered interest because of its profound effect on brain bioenergetics and neuronal survival in the pathophysiology of AD. Various therapeutic agents targeting the mitochondrial pathways associated with AD have been developed over the past decade. Although most of these agents are still early in the clinical development process, they are used to restore mitochondrial function, which provides an alternative therapeutic strategy that is likely to slow the progression of the disease. In this mini review, we will survey the AD-related mitochondrial pathways and their small-molecule modulators that have therapeutic potential. We will focus on recently reported examples, and also overview the current challenges and future perspectives of ongoing research. [BMB Reports 2020; 53(1): 47-55].


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Brain/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , 17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , 17-Hydroxysteroid Dehydrogenases/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/antagonists & inhibitors , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Animals , Disease Progression , Dynamins/metabolism , Humans , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Neurosteroids/chemistry , Neurosteroids/metabolism , Neurosteroids/pharmacology , Oxidative Stress/genetics , Oxidative Stress/physiology , Protein Kinases/chemistry , Protein Kinases/metabolism , Receptors, GABA/metabolism
10.
Drug Dev Res ; 80(8): 1013-1030, 2019 12.
Article in English | MEDLINE | ID: mdl-31823411

ABSTRACT

It is now firmly established that an important event in the formation of reperfusion injury of the heart is the opening of mitochondrial permeability transition pores (mPTPs), which changes the permeability of the mitochondria. mPTP opening results in the death of cardiomyocytes through activation of apoptosis and necroptosis. Experimental studies have shown that pharmacological inhibition of mPTP opening promotes the reduction in the infarct size and the suppression of apoptosis. Indeed, studies have shown the efficacy of mPTP inhibitors in animal models of myocardial reperfusion and isolated human myocardial trabeculae. However, clinical trials of cyclosporin A and TRO40303 have not provided a clear answer to the question of the effectiveness of mPTP inhibitors in patients with acute myocardial infarction. This article presents an analysis of possible approaches for the pharmacological regulation of mPTP during reperfusion injury of the heart.


Subject(s)
Cyclosporine/therapeutic use , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Myocardial Reperfusion Injury/drug therapy , Oximes/therapeutic use , Secosteroids/therapeutic use , Animals , Apoptosis/drug effects , Clinical Trials as Topic , Cyclosporine/pharmacology , Humans , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Oximes/pharmacology , Secosteroids/pharmacology
11.
J Recept Signal Transduct Res ; 39(4): 350-358, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31755334

ABSTRACT

Context: Although many studies have investigated the molecular mechanisms underlying hypoxia-related cardiomyocyte damage, the role of necrosis in cardiomyocyte death.Objective: The aim of our study is to explore the pathological role of nuclear receptor related 1 protein (NURR1) in regulating cardiomyocyte viability under hypoxia stress.Materials and methods: Cardiomyocyte was treated with hypoxia and siRNA against NURR1 was transfected into cardiomyocyte. Pathway agonist was used to activate the Mst1-JNK-mPTP pathway in cardiomyocyte.Results: In our study, the expression of NURR1 was rapidly increased in cardiomyocyte transfected with NURR1. Knockout of NURR1 could promote cardiomyocyte survival, reduce cell death and repress inflammation response. Mechanistically, NURR1 upregulation was associated with an activation of Mst1-JNK pathway and the latter promoted the mPTP opening in cardiomyocyte. Excessive mPTP opening was followed by cardiomyocyte necrosis and this effect could be reversed by NURR1 deletion. Besides, re-activation of Mst1-JNK pathway could abolish the protective effects of NURR1 deletion on cardiomyocytes, as evidenced by increased cell survival and decreased necrosis. Besides, re-activation of Mst1-JNK pathway also abolished NURR1 deletion-mediated mPTP opening.Conclusions: Hypoxia-mediated cardiomyocyte death is associated with NURR1 upregulation which contributes to the activation Mst1-JNK-mPTP pathways.


Subject(s)
Hepatocyte Growth Factor/antagonists & inhibitors , Hypoxia/physiopathology , Inflammation/prevention & control , MAP Kinase Kinase 4/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Myocytes, Cardiac/pathology , Necrosis , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Cells, Cultured , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Mice , Mitochondrial Permeability Transition Pore , Myocytes, Cardiac/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction
12.
ACS Comb Sci ; 21(12): 817-832, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31693340

ABSTRACT

When isolating binders from yeast displayed combinatorial libraries, a soluble, recombinantly expressed form of the target protein is typically utilized. As an alternative, we describe the use of target proteins displayed as surface fusions on magnetized yeast cells. In our strategy, the target protein is coexpressed on the yeast surface with an iron oxide binding protein; incubation of these yeast cells with iron oxide nanoparticles results in their magnetization. Subsequently, binder cells that interact with the magnetized target cells can be isolated using a magnet. Using a known binder-target pair with modest binding affinity (KD ≈ 400 nM), we showed that a binder present at low frequency (1 in 105) could be enriched more than 100-fold, in a single round of screening, suggesting feasibility of screening combinatorial libraries. Subsequently, we screened yeast display libraries of Sso7d and nanobody variants against yeast displayed targets to isolate binders specific to the cytosolic domain of the mitochondrial membrane protein TOM22 (KD ≈ 272-1934 nM) and the extracellular domain of the c-Kit receptor (KD ≈ 93 to KD > 2000 nM). Additional studies showed that the TOM22 binders identified using this approach could be used for the enrichment of mitochondria from cell lysates, thereby confirming binding to the native mitochondrial protein. The ease of expressing a membrane protein or a domain thereof as a yeast cell surface fusion-in contrast to recombinant soluble expression-makes the use of yeast-displayed targets particularly attractive. Therefore, we expect the use of magnetized yeast cell targets will enable efficient isolation of binders to membrane proteins.


Subject(s)
Ferric Compounds/pharmacology , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Nanoparticles/chemistry , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/drug effects , Small Molecule Libraries/pharmacology , Combinatorial Chemistry Techniques , Drug Evaluation, Preclinical , Ferric Compounds/chemistry , Magnetic Phenomena , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/isolation & purification , Mitochondrial Membrane Transport Proteins/metabolism , Protein Binding/drug effects , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/isolation & purification , Saccharomyces cerevisiae Proteins/metabolism , Small Molecule Libraries/chemistry
13.
J Physiol ; 597(24): 5879-5898, 2019 12.
Article in English | MEDLINE | ID: mdl-31631343

ABSTRACT

KEY POINTS: •Bile acids, ethanol and fatty acids affect pancreatic ductal fluid and bicarbonate secretion via mitochondrial damage, ATP depletion and calcium overload. •Pancreatitis-inducing factors open the membrane transition pore (mPTP) channel via cyclophilin D activation in acinar cells, causing calcium overload and cell death; genetic or pharmacological inhibition of mPTP improves the outcome of acute pancreatitis in animal models. •Here we show that genetic and pharmacological inhibition of mPTP protects mitochondrial homeostasis and cell function evoked by pancreatitis-inducing factors in pancreatic ductal cells. •The results also show that the novel cyclosporin A derivative NIM811 protects mitochondrial function in acinar and ductal cells, and it preserves bicarbonate transport mechanisms in pancreatic ductal cells. •We found that NIM811 is highly effective in different experimental pancreatitis models and has no side-effects. NIM811 is a highly suitable compound to be tested in clinical trials. ABSTRACT: Mitochondrial dysfunction plays a crucial role in the development of acute pancreatitis (AP); however, no compound is currently available with clinically acceptable effectiveness and safety. In this study, we investigated the effects of a novel mitochondrial transition pore inhibitor, N-methyl-4-isoleucine cyclosporin (NIM811), in AP. Pancreatic ductal and acinar cells were isolated by enzymatic digestion from Bl/6 mice. In vitro measurements were performed by confocal microscopy and microfluorometry. Preventative effects of pharmacological [cylosporin A (2 µm), NIM811 (2 µm)] or genetic (Ppif-/- /Cyp D KO) inhibition of the mitochondrial transition pore (mPTP) during the administration of either bile acids (BA) or ethanol + fatty acids (EtOH+FA) were examined. Toxicity of mPTP inhibition was investigated by detecting apoptosis and necrosis. In vivo effects of the most promising compound, NIM811 (5 or 10 mg kg-1 per os), were checked in three different AP models induced by either caerulein (10 × 50 µg kg-1 ), EtOH+FA (1.75 g kg-1 ethanol and 750 mg kg-1 palmitic acid) or 4% taurocholic acid (2 ml kg-1 ). Both genetic and pharmacological inhibition of Cyp D significantly prevented the toxic effects of BA and EtOH+FA by restoring mitochondrial membrane potential (Δψ) and preventing the loss of mitochondrial mass. In vivo experiments revealed that per os administration of NIM811 has a protective effect in AP by reducing oedema, necrosis, leukocyte infiltration and serum amylase level in AP models. Administration of NIM811 had no toxic effects. The novel mitochondrial transition pore inhibitor NIM811 thus seems to be an exceptionally good candidate compound for clinical trials in AP.


Subject(s)
Cyclosporine/therapeutic use , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Pancreatitis/drug therapy , Acinar Cells/drug effects , Acinar Cells/metabolism , Animals , Apoptosis , Bicarbonates/metabolism , Cells, Cultured , Cyclosporine/adverse effects , Cyclosporine/pharmacology , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Permeability Transition Pore , Pancreatic Ducts/drug effects , Pancreatic Ducts/metabolism
14.
ChemMedChem ; 14(20): 1771-1782, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31423734

ABSTRACT

Excessive mitochondrial matrix Ca2+ and oxidative stress leads to the opening of a high-conductance channel of the inner mitochondrial membrane referred to as the mitochondrial permeability transition pore (mtPTP). Because mtPTP opening can lead to cell death under diverse pathophysiological conditions, inhibitors of mtPTP are potential therapeutics for various human diseases. High throughput screening efforts led to the identification of a 3-carboxamide-5-phenol-isoxazole compounds as mtPTP inhibitors. While they showed nanomolar potency against mtPTP, they exhibited poor plasma stability, precluding their use in in vivo studies. Herein, we describe a series of structurally related analogues in which the core isoxazole was replaced with a triazole, which resulted in an improvement in plasma stability. These analogues were readily generated using the copper-catalyzed "click chemistry". One analogue, N-(5-chloro-2-methylphenyl)-1-(4-fluoro-3-hydroxyphenyl)-1H-1,2,3-triazole-4-carboxamide (TR001), was efficacious in a zebrafish model of muscular dystrophy that results from mtPTP dysfunction whereas the isoxazole isostere had minimal effect.


Subject(s)
Isoxazoles/pharmacology , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Muscular Dystrophies/drug therapy , Phenols/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Stability , HeLa Cells , High-Throughput Screening Assays , Humans , Isoxazoles/blood , Isoxazoles/chemistry , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Molecular Structure , Muscular Dystrophies/metabolism , Phenols/blood , Phenols/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured , Zebrafish
15.
Gastroenterology ; 157(5): 1368-1382, 2019 11.
Article in English | MEDLINE | ID: mdl-31336123

ABSTRACT

BACKGROUND & AIMS: Hepatic ischemia/reperfusion injury is a complication of liver surgery that involves mitochondrial dysfunction resulting from mitochondrial permeability transition pore (mPTP) opening. Cyclophilin D (PPIF or CypD) is a peptidyl-prolyl cis-trans isomerase that regulates mPTP opening in the inner mitochondrial membrane. We investigated whether and how recently created small-molecule inhibitors of CypD prevent opening of the mPTP in hepatocytes and the resulting effects in cell models and livers of mice undergoing ischemia/reperfusion injury. METHODS: We measured the activity of 9 small-molecule inhibitors of cyclophilins in an assay of CypD activity. The effects of the small-molecule CypD inhibitors or vehicle on mPTP opening were assessed by measuring mitochondrial swelling and calcium retention in isolated liver mitochondria from C57BL/6J (wild-type) and Ppif-/- (CypD knockout) mice and in primary mouse and human hepatocytes by fluorescence microscopy. We induced ischemia/reperfusion injury in livers of mice given a small-molecule CypD inhibitor or vehicle before and during reperfusion and collected samples of blood and liver for histologic analysis. RESULTS: The compounds inhibited peptidyl-prolyl isomerase activity (half maximal inhibitory concentration values, 0.2-16.2 µmol/L) and, as a result, calcium-induced mitochondrial swelling, by preventing mPTP opening (half maximal inhibitory concentration values, 1.4-132 µmol/L) in a concentration-dependent manner. The most potent inhibitor (C31) bound CypD with high affinity and inhibited swelling in mitochondria from livers of wild-type and Ppif-/- mice (indicating an additional, CypD-independent effect on mPTP opening) and in primary human and mouse hepatocytes. Administration of C31 in mice with ischemia/reperfusion injury before and during reperfusion restored hepatic calcium retention capacity and oxidative phosphorylation parameters and reduced liver damage compared with vehicle. CONCLUSIONS: Recently created small-molecule inhibitors of CypD reduced calcium-induced swelling in mitochondria from mouse and human liver tissues. Administration of these compounds to mice during ischemia/reperfusion restored hepatic calcium retention capacity and oxidative phosphorylation parameters and reduced liver damage. These compounds might be developed to protect patients from ischemia/reperfusion injury after liver surgery or for other hepatic or nonhepatic disorders related to abnormal mPTP opening.


Subject(s)
Enzyme Inhibitors/pharmacology , Liver Diseases/prevention & control , Liver/drug effects , Mitochondria, Liver/drug effects , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Peptidyl-Prolyl Isomerase F/antagonists & inhibitors , Reperfusion Injury/prevention & control , Animals , Calcium Signaling/drug effects , Cells, Cultured , Peptidyl-Prolyl Isomerase F/genetics , Peptidyl-Prolyl Isomerase F/metabolism , Cytoprotection , Disease Models, Animal , Humans , Liver/enzymology , Liver/pathology , Liver Diseases/enzymology , Liver Diseases/genetics , Liver Diseases/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Liver/enzymology , Mitochondria, Liver/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling/drug effects , Reperfusion Injury/enzymology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction
16.
Am J Physiol Endocrinol Metab ; 317(2): E269-E283, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31039005

ABSTRACT

Hydrogen sulfide (H2S) attenuates N-methyl-d-aspartate receptor-R1 (NMDA-R1) and mitigates diabetic renal damage; however, the molecular mechanism is not well known. Whereas NMDA-R1 facilitates Ca2+ permeability, H2S is known to inhibit L-type Ca2+ channel. High Ca2+ activates cyclophilin D (CypD), a gatekeeper protein of mitochondrial permeability transition pore (MPTP), thus facilitating molecular exchange between matrix and cytoplasm causing oxidative outburst and cell death. We tested the hypothesis of whether NMDA-R1 mediates Ca2+ influx causing CypD activation and MPTP opening leading to oxidative stress and renal injury in diabetes. We also tested whether H2S treatment blocks Ca2+ channel and thus inhibits CypD and MPTP opening to prevent renal damage. C57BL/6J and Akita (C57BL/6J-Ins2Akita) mice were treated without or with H2S donor GYY4137 (0.25 mg·kg-1·day-1 ip) for 8 wk. In vitro studies were performed using mouse glomerular endothelial cells. Results indicated that low levels of H2S and increased expression of NMDA-R1 in diabetes induced Ca2+ permeability, which was ameliorated by H2S treatment. We observed cytosolic Ca2+ influx in hyperglycemic (HG) condition along with mitochondrial-CypD activation, increased MPTP opening, and oxidative outburst, which were mitigated with H2S treatment. Renal injury biomarker KIM-1 was upregulated in HG conditions and normalized following H2S treatment. Inhibition of NMDA-R1 by pharmacological blocker MK-801 revealed similar results. We conclude that NMDA-R1-mediated Ca2+ influx in diabetes induces MPTP opening via CypD activation leading to increased oxidative stress and renal injury, and H2S protects diabetic kidney from injury by blocking mitochondrial Ca2+ permeability through NMDA-R1 pathway.


Subject(s)
Calcium/pharmacology , Diabetes Mellitus, Type 1/metabolism , Hydrogen Sulfide/pharmacology , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Cells, Cultured , Peptidyl-Prolyl Isomerase F/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Permeability Transition Pore
17.
Biochem J ; 476(11): 1585-1604, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31036718

ABSTRACT

Mitochondria play a central role in the maintenance of the naive state of embryonic stem cells. Many details of the mechanism remain to be fully elucidated. Solute carrier family 25 member 36 (Slc25a36) might regulate mitochondrial function through transporting pyrimidine nucleotides for mtDNA/RNA synthesis. Its physical role in this process remains unknown; however, Slc25a36 was recently found to be highly expressed in naive mouse embryonic stem cells (mESCs). Here, the function of Slc25a36 was characterized as a maintenance factor of mESCs pluripotency. Slc25a36 deficiency (via knockdown) has been demonstrated to result in mitochondrial dysfunction, which induces the differentiation of mESCs. The expression of key pluripotency markers (Pou5f1, Sox2, Nanog, and Utf1) decreased, while that of key TE genes (Cdx2, Gata3, and Hand1) increased. Cdx2-positive cells emerged in Slc25a36-deficient colonies under trophoblast stem cell culture conditions. As a result of Slc25a36 deficiency, mtDNA of knockdown cells declined, leading to impaired mitochondria with swollen morphology, decreased mitochondrial membrane potential, and low numbers. The key transcription regulators of mitochondrial biogenesis also decreased. These results indicate that mitochondrial dysfunction leads to an inability to support the pluripotency maintenance. Moreover, down-regulated glutathione metabolism and up-regulated focal adhesion reinforced and stabilized the process of differentiation by separately enhancing OCT4 degradation and promoting cell spread. This study improves the understanding of the function of Slc25a36, as well as the relationship of mitochondrial function with naive pluripotency maintenance and stem cell fate decision.


Subject(s)
Glutathione/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nucleotide Transport Proteins/metabolism , Animals , CDX2 Transcription Factor/metabolism , Cell Differentiation/genetics , Cells, Cultured , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Focal Adhesions , Gene Expression Regulation , Gene Knockdown Techniques , Mice , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Nucleotide Transport Proteins/antagonists & inhibitors , Nucleotide Transport Proteins/genetics , Octamer Transcription Factor-3/metabolism
19.
Future Med Chem ; 11(8): 857-867, 2019 04.
Article in English | MEDLINE | ID: mdl-30998114

ABSTRACT

Voltage-dependent anion channel isoform 3 (VDAC3), a channel in the mitochondrial outer membrane, has been suggested to play a role in the regulation of ATP transport and Ca2+ homeostasis. These processes are regarded as important for spermatozoa motility. Accordingly, in previous years, mutations in the VDAC3-encoding gene were detected in spermatozoa with low motility from infertile patients. Therefore, it can be assumed that these mutations would cause alteration of the structure and/or charge of the VDAC3 channel. The review is focused on current knowledge about contribution of VDAC3 activity to human spermatozoa motility and morphology. We also discuss the possibility of designing new molecules that could specifically block the VDAC3 channel and consequently act as male contraceptives.


Subject(s)
Contraceptive Agents, Male/pharmacology , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Spermatozoa/drug effects , Voltage-Dependent Anion Channels/antagonists & inhibitors , Voltage-Dependent Anion Channels/metabolism , Animals , Contraceptive Agents, Male/chemistry , Drug Design , Drug Discovery/methods , Humans , Male , Sperm Motility/drug effects , Spermatozoa/cytology , Spermatozoa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...