Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lefkothea C. Papadopoulou is active.

Publication


Featured researches published by Lefkothea C. Papadopoulou.


Nature Genetics | 1999

Fatal infantile cardioencephalomyopathy with COX deficiency and mutations in SCO2, a COX assembly gene.

Lefkothea C. Papadopoulou; Carolyn M. Sue; Mercy M. Davidson; Kurenai Tanji; Ichizo Nishino; James Sadlock; Sindu Krishna; Winsome F Walker; Jeanette Selby; D. Moira Glerum; Rudy Van Coster; Gilles Lyon; Emmanuel Scalais; Roger Lebel; Sara Shanske; Darryl C. De Vivo; Eduardo Bonilla; Michio Hirano; Salvatore DiMauro; Eric A. Schon

Mammalian cytochrome c oxidase (COX) catalyses the transfer of reducing equivalents from cytochrome c to molecular oxygen and pumps protons across the inner mitochondrial membrane. Mitochondrial DNA (mtDNA) encodes three COX subunits (I–III) and nuclear DNA (nDNA) encodes ten. In addition, ancillary proteins are required for the correct assembly and function of COX (refs 2, 3, 4, 5, 6). Although pathogenic mutations in mtDNA-encoded COX subunits have been described, no mutations in the nDNA-encoded subunits have been uncovered in any mendelian-inherited COX deficiency disorder. In yeast, two related COX assembly genes, SCO1 and SCO2 (for synthesis of cytochrome c oxidase), enable subunits I and II to be incorporated into the holoprotein. Here we have identified mutations in the human homologue, SCO2, in three unrelated infants with a newly recognized fatal cardioencephalomyopathy and COX deficiency. Immunohistochemical studies implied that the enzymatic deficiency, which was most severe in cardiac and skeletal muscle, was due to the loss of mtDNA-encoded COX subunits. The clinical phenotype caused by mutations in human SCO2 differs from that caused by mutations in SURF1, the only other known COX assembly gene associated with a human disease, Leigh syndrome.


Annals of Neurology | 2000

Differential features of patients with mutations in two COX assembly genes, SURF-1 and SCO2

Carolyn M. Sue; Charalampos Karadimas; Nicoletta Checcarelli; Kurenai Tanji; Lefkothea C. Papadopoulou; Francesco Pallotti; F. L. Guo; S. Shanske; Michio Hirano; D. C. De Vivo; R. Van Coster; Eduardo Bonilla; Salvatore DiMauro

We screened 41 patients with undiagnosed encephalomyopathies and cytochrome c oxidase (COX) deficiency for mutations in two COX assembly genes, SURF‐1 and SCO2; 6 patients had mutations in SURF‐1 and 3 had mutations in SCO2. All of the mutations in SURF‐1 were small‐scale rearrangements (deletions/insertions); 3 patients were homozygotes and the other 3 were compound heterozygotes. All patients with SCO2 mutations were compound heterozygotes for nonsense or missense mutations. All of the patients with mutations in SURF‐1 had Leigh syndrome, whereas the 3 patients with SCO2 mutations had a combination of encephalopathy and hypertrophic cardiomyopathy, and the neuropathology did not show the typical features of Leigh syndrome. In patients with SCO2 mutations, onset was earlier and the clinical course and progression to death more rapid than in patients with SURF‐1 mutations. In addition, biochemical and morphological studies showed that the COX deficiency was more severe in patients with SCO2 mutations. Immunohistochemical studies suggested that SURF‐1 mutations result in similarly reduced levels of mitochondrial‐encoded and nuclear‐encoded COX subunits, whereas SCO2 mutations affected mitochondrial‐encoded subunits to a greater degree. We conclude that patients with mutations in SURF‐1 and SCO2 genes have distinct phenotypes despite the common biochemical defect of COX activity. Ann Neurol 2000;47:589–595


Biochemical Pharmacology | 1999

Structural and Functional Impairment of Mitochondria in Adriamycin-Induced Cardiomyopathy in Mice: Suppression of Cytochrome c Oxidase II Gene Expression

Lefkothea C. Papadopoulou; George Theophilidis; George N. Thomopoulos; Asterios S. Tsiftsoglou

The use of adriamycin (ADR) in cancer chemotherapy has been limited due to its cumulative cardiovascular toxicity. Earlier observations that ADR interacts with mitochondrial cytochrome c oxidase (COX) and suppresses its enzyme activity led us to investigate ADRs action on the cardiovascular functions and heart mitochondrial morphology in Balb-c mice i.p. treated with ADR for several weeks. At various times during treatment, the animals were assessed for cardiovascular functions by electrocardiography and for heart tissue damage by electron microscopy. In parallel, total RNA was extracted from samples of dissected heart and analyzed by Northern blot hybridization to determine the steady-state level of three RNA transcripts encoded by the COXII, COXIII, and COXIV genes. Similarly, samples obtained from the liver of the same animals were analyzed for comparative studies. Our results indicated that 1) treatment of mice with ADR caused cardiovascular arrhythmias characterized by bradycardia, extension of ventricular depolarization time (tQRS), and failure of QRS at high concentrations (10-14 mg/kg body weight cumulative dose); 2) the heart mitochondria underwent swelling, fusion, dissolution, and/or disruption of mitochondrial cristae after several weeks of treatment. Such abnormalities were not observed in the mitochondria of liver tissue; and 3) among the three genes of COX enzyme examined, only COXII gene expression was suppressed by ADR treatment, mainly after 8 weeks in both heart and liver. Knowing that heart mitochondria represent almost 40% of heart muscle by weight, we conclude that the deteriorating effects of ADR on cardiovascular function involve mitochondrial structural and functional impairment.


American Journal of Human Genetics | 1997

The Fate of Human Sperm-Derived mtDNA in Somatic Cells

Giovanni Manfredi; Dominic Thyagarajan; Lefkothea C. Papadopoulou; Francesco Pallotti; Eric A. Schon

Inheritance of animal mtDNA is almost exclusively maternal, most likely because sperm-derived mitochondria are actively eliminated from the ovum, either at or soon after fertilization. How such elimination occurs is currently unknown. We asked whether similar behavior could be detected in somatic cells, by following the fate of mitochondria and mtDNAs after entry of human sperm into transformed cells containing mitochondria but lacking endogenous mtDNAs (rho0 cells). We found that a high proportion (10%-20%) of cells contained functioning sperm mitochondria soon after sperm entry. However, under selective conditions permitting only the survival of cells harboring functional mtDNAs, only approximately 1/10(5) cells containing sperm mitochondria survived and proliferated. These data imply that mitochondria in sperm can enter somatic cells relatively easily, but they also suggest that mechanisms exist to eliminate sperm-derived mtDNA from somatic cells, mechanisms perhaps similar to those presumed to operate in the fertilized oocyte.


Biochemical Pharmacology | 1996

Effects of hemin on apoptosis, suppression of cytochrome c oxidase gene expression, and bone-marrow toxicity induced by doxorubicin (adriamycin).

Lefkothea C. Papadopoulou; Asterios S. Tsiftsoglou

We have shown that hemin (iron-protoporphyrin IX) selectively counteracts doxorubicin (Adriamycin, ADR)-induced cytotoxicity on human leukemia K-562 cells by preventing ADR from inhibiting mitochondrial cytochrome c oxidase (COX), a novel target site for anthracyclines. Here, we investigated whether or not (a) treatment with ADR promotes apoptosis and represses the expression of two COX genes (one nuclear and one mitochondrial) in human K-562 cells in the absence and presence of hemin, and (b) injection of hemin preserves bone-marrow cellularity in ADR-myelosuppressed rats. Cultured K-562 cells were incubated with varying concentrations of ADR.HCl (0.2 microM to 5 microM) in the presence and absence of hemin (30 microM) and assessed for DNA degradation, as well as for expression of mitochondrial COXII and nuclear COXIV genes by RNA Northern blot hybridization analysis. In parallel, we investigated whether or not hemin injected i.p. in myelosuppressed rats affected ADR-induced bone-marrow cytotoxicity. These studies have shown the following: (a) ADR caused a dose- and time-dependent DNA fragmentation, characteristic of apoptosis, in K-562 cells; (b) hemin reduced the frequency of cell death caused by ADR: this effect was specific for ADR, because hemin failed to prevent apoptosis induced by methotrexate (MTX) in these cells; (c) ADR suppressed expression of COXIV and COXII genes, and exposure of ADR-treated K-562 cells to hemin did not reverse this suppression; and (d) i.p. injection of hemin in ADR-myelosuppressed rats improved bone-marrow cellularity, promoted colony formation (CFU-GM and CFU-F), and stromal cell outgrowth; moreover, hemin increased WBC counts depressed 12 days after ADR treatment. These studies indicate that hemin is a selective inhibitor of ADR-induced apoptosis of human leukemia cells and preserves bone-marrow cellularity in rats injected with ADR.


Journal of Inorganic Biochemistry | 2016

Synthesis, characterization and biological evaluation of 99m Tc/Re–tricarbonyl quinolone complexes

Theocharis E. Kydonaki; Evangelos Tsoukas; Filipa Mendes; Antonios G. Hatzidimitriou; António Paulo; Lefkothea C. Papadopoulou; Dionysia Papagiannopoulou; George Psomas

New rhenium(I) tricarbonyl complexes with the quinolone antimicrobial agents oxolinic acid (Hoxo) and enrofloxacin (Herx) and containing methanol, triphenylphosphine (PPh3) or imidazole (im) as unidentate co-ligands, were synthesized and characterized. The crystal structure of complex [Re(CO)3(oxo)(PPh3)]∙0.5MeOH was determined by X-ray crystallography. The deprotonated quinolone ligands are bound bidentately to rhenium(I) ion through the pyridone oxygen and a carboxylate oxygen. The binding of the rhenium complexes to calf-thymus DNA (CT DNA) was monitored by UV spectroscopy, viscosity measurements and competitive studies with ethidium bromide; intercalation was suggested as the most possible mode and the DNA-binding constants of the complexes were calculated. The rhenium complex [Re(CO)3(erx)(im)] was assayed for its topoisomerase IIα inhibition activity and was found to be active at 100μM concentration. The interaction of the rhenium complexes with human or bovine serum albumin was investigated by fluorescence emission spectroscopy (through the tryptophan quenching) and the corresponding binding constants were determined. The tracer complex [(99m)Tc(CO)3(erx)(im)] was synthesized and identified by comparative HPLC analysis with the rhenium analog. The (99m)Tc complex was found to be stable in solution. Upon injection in healthy mice, fast tissue clearance of the (99m)Tc complex was observed, while both renal and hepatobiliary excretion took place. Preliminary studies in human K-562 erythroleukemia cells showed cellular uptake of the (99m)Tc tracer with distribution primarily in the cytoplasm and the mitochondria and less in the nucleus. These preliminary results indicate that the quinolone (99m)Tc/Re complexes show promise to be further evaluated as imaging or therapeutic agents.


Gene | 2002

Differences in nuclear gene expression between cells containing monomer and dimer mitochondrial genomes

Kim M. Clark; Timothy A. Brown; Mercy M. Davidson; Lefkothea C. Papadopoulou; David A. Clayton

It is known that point mutations and rearrangements (deletions and duplications) of mammalian mitochondrial DNA (mtDNA) can result in mitochondrial dysfunction and human disease. Very little attention has been paid to mtDNA circular dimers (a complex form consisting of two genomes joined head-to-tail) despite their close association with human neoplasia. MtDNA dimers are frequently found in human leukemia, but the clinical relevance of their presence remains unknown. To begin to investigate the role of circular dimer mtDNA in the tumorigenic phenotype, we have created isogenic cell lines containing monomer and dimer mitochondrial genomes and compared the respective nuclear mRNA expression using Affymetrix gene array analysis. Surprisingly, a large number of nuclear gene changes were observed, with one of the largest category of genes being associated with remodeling of the cell surface and extracellular matrix. Since cell growth, migration, apoptosis, and many other cellular processes are influenced by signals initiating from the cell surface, the changes associated with the presence of mtDNA dimers could lead to significant alterations in tumorigenic potential and/or progression.


Biochimica et Biophysica Acta | 2010

Intracellular delivery of full length recombinant human mitochondrial L-Sco2 protein into the mitochondria of permanent cell lines and SCO2 deficient patient's primary cells

Parthena F. Foltopoulou; Asterios S. Tsiftsoglou; Ioannis D. Bonovolias; Alexandra T. Ingendoh; Lefkothea C. Papadopoulou

Mutations in human SCO2 gene, encoding the mitochondrial inner membrane Sco2 protein, have been found to be responsible for fatal infantile cardioencephalomyopathy and cytochrome c oxidase (COX) deficiency. One potentially fruitful therapeutic approach for this mitochondrial disorder should be considered the production of human recombinant full length L-Sco2 protein and its deliberate transduction into the mitochondria. Recombinant L-Sco2 protein, fused with TAT, a Protein Transduction Domain (PTD), was produced in bacteria and purified from inclusion bodies (IBs). Following solubilisation with l-arginine, this fusion L-Sco2 protein was transduced in cultured mammalian cells of different origin (U-87 MG, T24, K-562, and patients primary fibroblasts) and assessed for stability, transduction into mitochondria, processing and impact on recovery of COX activity. Our results indicate that: a) l-Arg solution was effective in solubilising recombinant fusion L-Sco2 protein, derived from IBs; b) fusion L-Sco2 protein was delivered successfully via a time- and concentration-dependent process into the mitochondria of human U-87 MG and T24 cells; c) fusion L-Sco2 protein was also transduced in human K-562 cells, transiently depleted of SCO2 transcripts and thus COX deficient; transduction of this fusion protein led to partial recovery of COX activity in such cells; d) [(35)S]Methionine-labelled fusion L-Sco2 protein, produced in a cell free transcription/translation system and incubated with intact isolated mitochondria derived from K-562 cells, was efficiently processed to yield the corresponding mature Sco2 protein, thus justifying the potential of the transduced fusion L-Sco2 protein to successfully activate COX holoenzyme; and finally, e) recombinant fusion L-Sco2 protein was successfully transduced into the mitochondria of primary fibroblasts derived from SCO2/COX deficient patient and facilitated recovery of COX activity. These findings provide the rationale of delivering recombinant proteins via PTD technology as a model for therapeutic approach of mitochondrial disorders.


Pharmaceutical Research | 2011

Transduction of Human Recombinant Proteins into Mitochondria as a Protein Therapeutic Approach for Mitochondrial Disorders

Lefkothea C. Papadopoulou; Asterios S. Tsiftsoglou

ABSTRACTProtein therapy is considered an alternative approach to gene therapy for treatment of genetic-metabolic disorders. Human protein therapeutics (PTs), developed via recombinant DNA technology and used for the treatment of these illnesses, act upon membrane-bound receptors to achieve their pharmacological response. On the contrary, proteins that normally act inside the cells cannot be developed as PTs in the conventional way, since they are not able to “cross” the plasma membrane. Furthermore, in mitochondrial disorders, attributed either to depleted or malfunctioned mitochondrial proteins, PTs should also have to reach the subcellular mitochondria to exert their therapeutic potential. Nowadays, there is no effective therapy for mitochondrial disorders. The development of PTs, however, via the Protein Transduction Domain (PTD) technology offered new opportunities for the deliberate delivery of human recombinant proteins inside eukaryotic subcellular organelles. To this end, mitochondrial disorders could be clinically encountered with the delivery of human mitochondrial proteins (engineered via recombinant DNA and PTD technologies) at specific intramitochondrial sites to exert their function. Overall, PTD-mediated Protein Replacement Therapy emerges as a suitable model system for the therapeutic approach for mitochondrial disorders.


Pharmaceuticals | 2013

The potential role of cell penetrating peptides in the intracellular delivery of proteins for therapy of erythroid related disorders.

Lefkothea C. Papadopoulou; Asterios S. Tsiftsoglou

The erythroid related disorders (ERDs) represent a large group of hematological diseases, which in most cases are attributed either to the deficiency or malfunction of biosynthetic enzymes or oxygen transport proteins. Current treatments for these disorders include histo-compatible erythrocyte transfusions or allogeneic hematopoietic stem cell (HSC) transplantation. Gene therapy delivered via suitable viral vectors or genetically modified HSCs have been under way. Protein Transduction Domain (PTD) technology has allowed the production and intracellular delivery of recombinant therapeutic proteins, bearing Cell Penetrating Peptides (CPPs), into a variety of mammalian cells. Remarkable progress in the field of protein transduction leads to the development of novel protein therapeutics (CPP-mediated PTs) for the treatment of monogenetic and/or metabolic disorders. The “concept” developed in this paper is the intracellular protein delivery made possible via the PTD technology as a novel therapeutic intervention for treatment of ERDs. This can be achieved via four stages including: (i) the production of genetically engineered human CPP-mediated PT of interest, since the corresponding native protein either is missing or is mutated in the erythroid progenitor cell (ErPCs) or mature erythrocytes of patients; (ii) isolation of target cells from the peripheral blood of the selected patients; (iii) ex vivo transduction of cells with the CPP-mediated PT of interest; and (iv) re-administration of the successfully transduced cells back into the same patients.

Collaboration


Dive into the Lefkothea C. Papadopoulou's collaboration.

Top Co-Authors

Avatar

Asterios S. Tsiftsoglou

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar

Parthena F. Foltopoulou

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar

Ioannis D. Bonovolias

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar

Carolyn M. Sue

Royal North Shore Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric A. Schon

Columbia University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michio Hirano

Columbia University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge