Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cells ; 13(12)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38920700

ABSTRACT

Cancer accounted for 10 million deaths in 2020, nearly one in every six deaths annually. Despite advancements, the contemporary clinical management of human neoplasms faces a number of challenges. Surgical removal of tumor tissues is often not possible technically, while radiation and chemotherapy pose the risk of damaging healthy cells, tissues, and organs, presenting complex clinical challenges. These require a paradigm shift in developing new therapeutic modalities moving towards a more personalized and targeted approach. The tumor-agnostic philosophy, one of these new modalities, focuses on characteristic molecular signatures of transformed cells independently of their traditional histopathological classification. These include commonly occurring DNA aberrations in cancer cells, shared metabolic features of their homeostasis or immune evasion measures of the tumor tissues. The first dedicated, FDA-approved tumor-agnostic agent's profound progression-free survival of 78% in mismatch repair-deficient colorectal cancer paved the way for the accelerated FDA approvals of novel tumor-agnostic therapeutic compounds. Here, we review the historical background, current status, and future perspectives of this new era of clinical oncology.


Subject(s)
Neoplasms , Humans , Neoplasms/therapy , Neoplasms/genetics , Neoplasms/pathology , Precision Medicine
2.
Pharmaceutics ; 16(3)2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38543275

ABSTRACT

Cardiovascular disease constitutes a noteworthy public health challenge characterized by a pronounced incidence, frequency, and mortality rate, particularly impacting specific demographic groups, and imposing a substantial burden on the healthcare infrastructure. Certain risk factors, such as age, gender, and smoking, contribute to the prevalence of fatal cardiovascular disease, highlighting the need for targeted interventions. Current challenges in clinical practice involve medication complexities, the lack of a systematic decision-making approach, and prevalent drug therapy problems. Stem cell-derived extracellular vesicles stand as versatile entities with a unique molecular fingerprint, holding significant therapeutic potential across a spectrum of applications, particularly in the realm of cardio-protection. Their lipid, protein, and nucleic acid compositions, coupled with their multifaceted functions, underscore their role as promising mediators in regenerative medicine and pave the way for further exploration of their intricate contributions to cellular physiology and pathology. Here, we overview our current understanding of the possible role of stem cell-derived extracellular vesicles in the clinical management of human cardiovascular pathologies.

3.
Biology (Basel) ; 12(12)2023 Nov 24.
Article in English | MEDLINE | ID: mdl-38132289

ABSTRACT

Based on recent advances in research of chronic inflammatory conditions, there is a growing body of evidence that suggests a close correlation between the microbiota of the gastrointestinal tract and the physiologic activity of the immune system. This raises the idea that disturbances of the GI ecosystem contribute to the unfolding of chronic diseases including neurodegenerative pathologies. Here, we overview our current understanding on the putative interaction between the gut microbiota and the immune system from the aspect of multiple sclerosis, one of the autoimmune conditions accompanied by severe chronic neuroinflammation that affects millions of people worldwide.

4.
J Alzheimers Dis ; 86(1): 343-364, 2022.
Article in English | MEDLINE | ID: mdl-35034897

ABSTRACT

BACKGROUND: The effects of the key pathogens and virulence factors associated with gum disease such as Porphyromonas gingivalis (P. gingivalis) on the central nervous system is of great interest with respect to development of neuropathologies and hence therapeutics and preventative strategies. Chronic infections and associated inflammation are known to weaken the first line of defense for the brain, the blood-brain barrier (BBB). OBJECTIVE: The focus of this study is to utilize an established human in vitro BBB model to evaluate the effects of P. gingivalis virulence factors lipopolysaccharide (LPS) and outer membrane vesicles (OMVs) on a primary-derived human model representing the neurovascular unit of the BBB. METHODS: Changes to the integrity of the BBB after application of P. gingivalis LPS and OMVs were investigated and correlated with transport of LPS. Additionally, the effect of P. gingivalis LPS and OMVs on human brain microvascular endothelial cells in monolayer was evaluated using immunofluorescence microscopy. RESULTS: The integrity of the BBB model was weakened by application of P. gingivalis LPS and OMVs, as measured by a decrease in electrical resistance and a recovery deficit was seen in comparison to the controls. Application of P. gingivalis OMVs to a monoculture of human brain microvascular endothelial cells showed disruption of the tight junction zona occludens protein (ZO-1) compared to controls. CONCLUSION: These findings show that the integrity of tight junctions of the human BBB could be weakened by association with P. gingivalis virulence factors LPS and OMVs containing proteolytic enzymes (gingipains).


Subject(s)
Lipopolysaccharides , Porphyromonas gingivalis , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Humans , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Permeability , Tight Junction Proteins/metabolism , Virulence Factors
5.
Cells ; 10(9)2021 08 26.
Article in English | MEDLINE | ID: mdl-34571848

ABSTRACT

Cellular therapy is a promising tool of human medicine to successfully treat complex and challenging pathologies such as cardiovascular diseases or chronic inflammatory conditions. Bone marrow-derived mesenchymal stromal cells (BMSCs) are in the limelight of these efforts, initially, trying to exploit their natural properties by direct transplantation. Extensive research on the therapeutic use of BMSCs shed light on a number of key aspects of BMSC physiology including the importance of oxygen in the control of BMSC phenotype. These efforts also led to a growing number of evidence indicating that the beneficial therapeutic effects of BMSCs can be mediated by BMSC-secreted agents. Further investigations revealed that BMSC-excreted extracellular vesicles could mediate the potentially therapeutic effects of BMSCs. Here, we review our current understanding of the relationship between low oxygen conditions and the effects of BMSC-secreted extracellular vesicles focusing on the possible medical relevance of this interplay.


Subject(s)
Extracellular Vesicles/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Animals , Bone Marrow Cells/cytology , Cell Differentiation/physiology , Cell Hypoxia/physiology , Cell- and Tissue-Based Therapy/methods , Extracellular Vesicles/transplantation , Humans , Mesenchymal Stem Cells/physiology , Oxygen/metabolism
6.
Stem Cells Int ; 2021: 8899756, 2021.
Article in English | MEDLINE | ID: mdl-33519938

ABSTRACT

Stem cell-based cellular therapy is a promising tool for the treatment of pathological conditions with underlying severe tissue damage or malfunction like in chronic cardiovascular, musculoskeletal, or inflammatory conditions. One of the biggest technical challenges of the use of natural stem cells, however, is the prevention of their premature senescence during therapeutical manipulations. Culturing stem cells under hypoxic conditions is believed to be a possible route to fulfill this goal. Here, we review current literature data on the effects of hypoxia on bone marrow-derived mesenchymal stromal cells, one of the most popular tools of practical cellular therapy, in the context of their senescence.

7.
Stem Cells Int ; 2019: 2509606, 2019.
Article in English | MEDLINE | ID: mdl-31687031

ABSTRACT

The therapeutic repertoire for life-threatening inflammatory conditions like sepsis, graft-versus-host reactions, or colitis is very limited in current clinical practice and, together with chronic ones, like the osteoarthritis, presents growing economic burden in developed countries. This urges the development of more efficient therapeutic modalities like the mesenchymal stem cell-based approaches. Despite the encouraging in vivo data, however, clinical trials delivered ambiguous results. Since one of the typical features of inflamed tissues is decreased oxygenation, the success of cellular therapy in inflammatory pathologies seems to be affected by the impact of oxygen depletion on transplanted cells. Here, we examine our current knowledge on the effect of hypoxia on the physiology of bone marrow-derived mesenchymal stromal cells, one of the most popular tools of practical cellular therapy, in the context of their immune-modulatory capacity.

8.
Front Physiol ; 10: 538, 2019.
Article in English | MEDLINE | ID: mdl-31133874

ABSTRACT

PARP-1 inhibition has recently been employed in both mono- and combination therapies in various malignancies including melanoma with both promising and contradicting results reported. Although deeper understanding of the underlying molecular mechanisms may help improving clinical modalities, the complex cellular effects of PARP inhibitors make disentangling of the mechanisms involved in combination therapies difficult. Here, we used two cytostatic agents used in melanoma therapies in combination with PARP inhibition to have an insight into cellular events using the B16F10 melanoma model. We found that, when used in combination with cisplatin or temozolomide, pharmacologic blockade of PARP-1 by PJ34 augmented the DNA-damaging and cytotoxic effects of both alkylating compounds. Interestingly, however, this synergism unfolds relatively slowly and is preceded by molecular events that are traditionally believed to support cell survival including the stabilization of mitochondrial membrane potential and morphology. Our data indicate that the PARP inhibitor PJ34 has, apparently, opposing effects on the mitochondrial structure and cell survival. While, initially, it stimulates mitochondrial fusion and hyperpolarization, hallmarks of mitochondrial protection, it enhances the cytotoxic effects of alkylating agents at later stages. These findings may contribute to the optimization of PARP inhibitor-based antineoplastic modalities.

9.
J Immunol ; 202(5): 1521-1530, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30700584

ABSTRACT

Hypoxia is a common and prominent feature of the microenvironment at sites of bacteria-associated inflammation in inflammatory bowel disease. The prolyl-hydroxylases (PHD1/2/3) and the asparaginyl-hydroxylase factor-inhibiting HIF are oxygen-sensing enzymes that regulate adaptive responses to hypoxia through controlling the activity of HIF and NF-κB-dependent transcriptional pathways. Previous studies have demonstrated that the pan-hydroxylase inhibitor dimethyloxalylglycine (DMOG) is effective in the alleviation of inflammation in preclinical models of inflammatory bowel disease, at least in part, through suppression of IL-1ß-induced NF-κB activity. TLR-dependent signaling in immune cells, such as monocytes, which is important in bacteria-driven inflammation, shares a signaling pathway with IL-1ß. In studies into the effect of pharmacologic hydroxylase inhibition on TLR-induced inflammation in monocytes, we found that DMOG selectively triggers cell death in cultured THP-1 cells and primary human monocytes at concentrations well tolerated in other cell types. DMOG-induced apoptosis was independent of increased caspase-3/7 activity but was accompanied by reduced expression of the inhibitor of apoptosis protein 1 (cIAP1). Based on these data, we hypothesize that pharmacologic inhibition of the HIF-hydroxylases selectively targets monocytes for cell death and that this may contribute to the anti-inflammatory activity of HIF-hydroxylase inhibitors.


Subject(s)
Amino Acids, Dicarboxylic/pharmacology , Inflammation/drug therapy , Mixed Function Oxygenases/antagonists & inhibitors , Monocytes/drug effects , Prolyl-Hydroxylase Inhibitors/pharmacology , Cell Death/drug effects , Cell Death/immunology , Cells, Cultured , HEK293 Cells , Humans , Inflammation/immunology , Inflammation/metabolism , Mixed Function Oxygenases/immunology , Mixed Function Oxygenases/metabolism , Monocytes/immunology , Monocytes/metabolism
10.
Oncotarget ; 8(30): 50221-50239, 2017 Jul 25.
Article in English | MEDLINE | ID: mdl-28430591

ABSTRACT

Poly(ADP-ribose) polymerases are a family of DNA-dependent nuclear enzymes catalyzing the transfer of ADP-ribose moieties from cellular nicotinamide-adenine-dinucleotide to a variety of target proteins. Although they have been considered as resident nuclear elements of the DNA repair machinery, recent works revealed a more intricate physiologic role of poly(ADP-ribose) polymerases with numerous extranuclear activities. Indeed, poly(ADP-ribose) polymerases participate in fundamental cellular processes like chromatin remodelling, transcription or regulation of the cell-cycle. These new insight into the physiologic roles of poly(ADP-ribose) polymerases widens the range of human pathologies in which pharmacologic inhibition of these enzymes might have a therapeutic potential. Here, we overview our current knowledge on extranuclear functions of poly(ADP-ribose) polymerases with a particular focus on the mitochondrial ones and discuss potential fields of future clinical applications.


Subject(s)
Mitochondria/metabolism , Poly(ADP-ribose) Polymerases/genetics , Humans , Poly(ADP-ribose) Polymerases/metabolism , Signal Transduction
11.
Sci Rep ; 6: 31355, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27531581

ABSTRACT

Cellular exposure to hypoxia results in altered gene expression in a range of physiologic and pathophysiologic states. Discrete cohorts of genes can be either up- or down-regulated in response to hypoxia. While the Hypoxia-Inducible Factor (HIF) is the primary driver of hypoxia-induced adaptive gene expression, less is known about the signalling mechanisms regulating hypoxia-dependent gene repression. Using RNA-seq, we demonstrate that equivalent numbers of genes are induced and repressed in human embryonic kidney (HEK293) cells. We demonstrate that nuclear localization of the Repressor Element 1-Silencing Transcription factor (REST) is induced in hypoxia and that REST is responsible for regulating approximately 20% of the hypoxia-repressed genes. Using chromatin immunoprecipitation assays we demonstrate that REST-dependent gene repression is at least in part mediated by direct binding to the promoters of target genes. Based on these data, we propose that REST is a key mediator of gene repression in hypoxia.


Subject(s)
Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Analysis, RNA/methods , Transcription, Genetic , Cell Hypoxia , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , HEK293 Cells , Humans , Promoter Regions, Genetic , Signal Transduction
12.
Biochem Biophys Res Commun ; 474(3): 579-586, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27130823

ABSTRACT

Hepatocyte death is an important contributing factor in a number of diseases of the liver. PHD1 confers hypoxic sensitivity upon transcription factors including the hypoxia inducible factor (HIF) and nuclear factor-kappaB (NF-κB). Reduced PHD1 activity is linked to decreased apoptosis. Here, we investigated the underlying mechanism(s) in hepatocytes. Basal NF-κB activity was elevated in PHD1(-/-) hepatocytes compared to wild type controls. ChIP-seq analysis confirmed enhanced binding of NF-κB to chromatin in regions proximal to the promoters of genes involved in the regulation of apoptosis. Inhibition of NF-κB (but not knock-out of HIF-1 or HIF-2) reversed the anti-apoptotic effects of pharmacologic hydroxylase inhibition. We hypothesize that PHD1 inhibition leads to altered expression of NF-κB-dependent genes resulting in reduced apoptosis. This study provides new information relating to the possible mechanism of therapeutic action of hydroxylase inhibitors that has been reported in pre-clinical models of intestinal and hepatic disease.


Subject(s)
Apoptosis/physiology , Hepatocytes/cytology , Hepatocytes/physiology , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , NF-kappa B/metabolism , Procollagen-Proline Dioxygenase/metabolism , Animals , Cell Hypoxia/physiology , Cell Line , Gene Expression Regulation, Enzymologic/physiology , HEK293 Cells , Humans , Mice
13.
J Mol Med (Berl) ; 94(4): 377-90, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26821588

ABSTRACT

Hypoxia is a common micro-environmental stress which is experienced by cells during a range of physiologic and pathophysiologic processes. The identification of the hypoxia-inducible factor (HIF) as the master regulator of the transcriptional response to hypoxia transformed our understanding of the mechanism underpinning the hypoxic response at the molecular level and identified HIF as a potentially important new therapeutic target. It has recently become clear that multiple levels of regulatory control exert influence on the HIF pathway giving the response a complex and dynamic activity profile. These include positive and negative feedback loops within the HIF pathway as well as multiple levels of crosstalk with other signaling pathways. The emerging model reflects a multi-level regulatory network that affects multiple aspects of the physiologic response to hypoxia including proliferation, apoptosis, and differentiation. Understanding the interplay between the molecular mechanisms involved in the dynamic regulation of the HIF pathway at a systems level is critically important in defining new appropriate therapeutic targets for human diseases including ischemia, cancer, and chronic inflammation. Here, we review our current knowledge of the regulatory circuits which exert influence over the HIF response and give examples of in silico model-based predictions of the dynamic behaviour of this system.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Models, Biological , Signal Transduction , Systems Biology , Animals , Carrier Proteins/metabolism , Feedback, Physiological , Gene Expression Regulation , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Isoenzymes , MAP Kinase Signaling System , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Prolyl Hydroxylases/metabolism , Protein Binding , Systems Biology/methods
14.
Stem Cell Res ; 12(3): 646-58, 2014 May.
Article in English | MEDLINE | ID: mdl-24667757

ABSTRACT

Human bone marrow stromal cells (hBMSCs, also known as bone marrow-derived mesenchymal stem cells) are promising tools for the cellular therapy of human pathologies related to various forms of hypoxia. Although the current concepts of their clinical use include the expansion of hBMSC in standard cell culture conditions, the effect of the mitogen-driven ex vivo expansion on the adaptation to the hypoxic environment is unknown. Here, we provide data that the basic fibroblast growth factor (FGF2) enhances the induction of a wide range of hypoxia-related adaptive genes in hypoxic hBMSCs. We identified that the FGF2 signal is transmitted by the ERK pathway similar to that of hypoxia that also utilises the distal elements of the same signalling machinery including the extracellular signal-regulated kinase 1/2 (ERK1/2) and mitogen-activated protein kinase kinases (MEK1/2) in hBMSCs. We found that the simultaneous activation of ERK1/2 by FGF2 and hypoxia transforms the activation dynamics from oscillatory into sustained one. Activated ERKs co-localise with stabilised hypoxia inducible factor-1α (HIF-1α) followed by the reduction of its nuclear mobility as well as increased DNA binding capacity leading to the up-regulation of hypoxia-adaptive genes. Our findings indicate that the status of the ERK pathway has significant impacts on the molecular adaptation of hBMSCs to the hypoxic milieu.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MAP Kinase Signaling System , Mesenchymal Stem Cells/metabolism , Cell Hypoxia , Cell Proliferation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mesenchymal Stem Cells/cytology , Oxygen/metabolism , Promoter Regions, Genetic , Protein Binding , Up-Regulation
15.
World J Biol Psychiatry ; 11(4): 667-76, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20334575

ABSTRACT

OBJECTIVES: Atypical antipsychotic agents constitute one therapeutic approach for bipolar disorder. Since disease course and outcome are variable, further studies are needed to complement limited data supportive of clinical decisions at treatment initiation. METHODS: This 12-month, prospective, observational study investigated factors associated with symptomatic remission (total YMRS score < or =12) and full clinical recovery (sustained reduction in CGI-BP-S overall score) in bipolar disorder during treatment with atypical antipsychotics (predominantly olanzapine, risperidone and quetiapine; alone or in combination with a psychotropic such as lithium or valproate) in actual clinical practice. RESULTS: Amongst 872 enrolled and eligible patients, rates of symptomatic remission and full clinical recovery at 12 months were 93.0 and 78.5%, respectively. Of the baseline factors significantly (P< or =0.05) associated with symptomatic remission, the following categories were positively associated with a higher chance of symptomatic remission: Caucasian ethnicity; higher CGI-BP-S scores; family-dependent living; a previous manic episode; 1, 2 or > or =5 social activities; no work impairment; and being neither satisfied nor dissatisfied with life. Outpatient treatment and historically longer periods of mania were significantly positively associated with full clinical recovery. CONCLUSIONS: While clinical status may also be associated with longer-term remission and recovery, factors relating to social functioning and quality of life are easily assessed and potentially modifiable. Such knowledge may aid physicians' clinical decisions regarding patients with bipolar mania treated with atypical antipsychotics.


Subject(s)
Antipsychotic Agents/therapeutic use , Bipolar Disorder/drug therapy , Ethnicity/statistics & numerical data , Adult , Benzodiazepines/therapeutic use , Bipolar Disorder/epidemiology , Bipolar Disorder/psychology , Comorbidity , Diagnostic and Statistical Manual of Mental Disorders , Dibenzothiazepines/therapeutic use , Female , Humans , Lithium Carbonate/therapeutic use , Male , Observation , Olanzapine , Personality Disorders/epidemiology , Prospective Studies , Quetiapine Fumarate , Remission Induction , Risperidone/therapeutic use , Severity of Illness Index , Substance-Related Disorders/epidemiology , Valproic Acid/therapeutic use
16.
Orv Hetil ; 148(43): 2019-26, 2007 Oct 28.
Article in Hungarian | MEDLINE | ID: mdl-17947194

ABSTRACT

The introduction of new therapeutical approaches to the medical practice has never been an easy task. On the one hand, modern therapies have to fulfil the strict requirements of the industry, on the other, patients and therapists have usually high expectations towards them. Thus, the public's initial euphoric emotions can quickly turn into pessimism following the experience of the first complications and difficulties of the new therapeutic method. Regarding oncolytic virotherapy, the emerging troubles and the lack of complete success upon the first clinical trials also led to the rejection of this promising new approach to the treatment of cancer. In the last few decades, however, due to the advances in molecular and cell biology, oncolytic virotherapy has been put in the lime-light again, and some of the newly developed virotherapeutic agents have been tested in various clinical trials all around the world. This review focuses on the current situation of the field and summarizes the available results.


Subject(s)
Neoplasms/drug therapy , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Tumor Virus Infections/complications , Adenoviruses, Human/genetics , Animals , Humans , Neoplasms/virology , Newcastle disease virus/genetics , Oncolytic Virotherapy/methods , Poliovirus/genetics , Reoviridae/genetics , Simplexvirus/genetics , Tumor Virus Infections/virology , Vaccinia virus/genetics
17.
J Virol ; 81(6): 2817-30, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17215292

ABSTRACT

While Newcastle disease virus (NDV) causes serious infections in birds, it is apparently nonpathogenic in mammalian species, including humans. Previous observations and small-scale clinical trials indicated that NDV exerts oncolytic effects. Isolates of NDV were found to have selective affinity to transformed cells. We previously showed that the attenuated NDV strain MTH-68/H causes apoptotic cell death in cultures of PC12 rat pheochromocytoma cells. The aim of the present study was to extend MTH-68/H cytotoxicity testing with human tumor cell lines and to analyze certain biochemical aspects of its oncolytic effect. MTH-68/H was found to be able to kill a wide range of transformed cells by apoptosis. While caspase-8 and caspase-9 are not involved in MTH-68/H-induced apoptosis, activation of caspase-3 and caspase-12 was detected in virus-infected PC12 cells. A human glioblastoma cell line with repressible expression of the p53 protein did not show any difference in MTH-68/H sensitivity in its p53-expressing and p53-depleted states, indicating that the apoptotic process induced by MTH-68/H does not depend on p53. Apoptosis was accompanied by virus replication in two tumor cell lines tested (PC12 cells and HeLa human cervical cells), and signs of endoplasmic reticulum stress (phosphorylation of protein kinase R-like endoplasmic reticulum kinase and eIF2alpha) were also detected in transformed cells. In contrast, proliferation of nontransformed mouse and rat fibroblast cell lines and human primary fibroblasts was not affected by MTH-68/H treatment. MTH-68/H thus selectively kills tumor cell cultures by inducing endoplasmic reticulum stress leading to p53-independent apoptotic cell death.


Subject(s)
Cell Transformation, Viral/physiology , Endoplasmic Reticulum/physiology , Newcastle disease virus/physiology , Oxidative Stress/physiology , Tumor Suppressor Protein p53/physiology , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis , Carcinoma/pathology , Caspase 12/metabolism , Caspase 3/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation , Enzyme Activation , Female , Glioblastoma/pathology , HCT116 Cells , HT29 Cells , HeLa Cells , Humans , Newcastle disease virus/genetics , PC12 Cells , Pancreatic Neoplasms/pathology , Rats , Tetracycline/pharmacology , Virus Replication
18.
J Cell Biochem ; 99(5): 1431-41, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16817227

ABSTRACT

PC12 rat phaeochromocytoma cells show neuronal differentiation upon NGF treatment. NGF induces prolonged activation of the Ras/Raf/MEK/ERK pathway in which the 42/44 kDa mitogen-activated protein kinases (MAPKs), ERK 1 and 2 are thought to be the key mediators of the differentiation signals. Activation of ERKs leads to the increased transcription of early response genes resulting in cell cycle arrest. Upon NGF treatment the p53 protein, the most commonly mutated tumor suppressor in human cancers, translocates to the nucleus and may play a role in the mediation of NGF-induced cell cycle arrest and neuronal differentiation. Here we demonstrate that in PC12 cells expressing both wild-type and V143A mutant p53 proteins (p143p53PC12 cells), p53-mediated biological responses are critically influenced. p143p53PC12 cells are not able to cease their proliferation and begin their neuronal differentiation program upon NGF treatment. The presence of mutant p53 also reduces the DNA-binding activity of endogenous p53 and disturbs the regulatory machinery of p53 including both the phosphorylation of ERK 1/2, p38 and SAPK/JNK MAP kinases and itself.


Subject(s)
Cell Differentiation , Cell Proliferation , PC12 Cells , Tumor Suppressor Protein p53/metabolism , Animals , Enhancer Elements, Genetic , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/metabolism , Neurons/cytology , Neurons/physiology , Phosphorylation , Rats , Tumor Suppressor Protein p53/genetics
19.
Neuropsychopharmacol Hung ; 7(3): 146-56, 2005 Sep.
Article in Hungarian | MEDLINE | ID: mdl-16493879

ABSTRACT

AIM OF THE STUDY: One of the most frequent causes of temporary or permanent diminution of work productivity is depression. The complete recovery of depression means fully restored creativity, work production and life satisfaction. Antidepressive treatment is regarded effective if besides symptom remission, the original functions and roles are fully restored, i.e. life satisfaction significantly improves. The aim of this study was to assess the improvement of work productivity and life satisfaction of paroxetin treated depressives. METHODS: The study was a 52 weeks lasting open, multicentric, self-controlling survey with the inclusion of 1407 paroxetin treated depressed patients. Apart from recording demographic data, comorbidities, course of the illness, and scales measuring the severity of the illness (HAMD, CGI-S, CGI-I), scales indicating the impairment of work productivity (SAS, SDS) and life satisfaction (LS) were also processed. RESULTS: Besides the improvement of the depressive symptoms (HAMD scores decreased from 22.6 to 3.8; and CGI-S scores from 4.7 to 1.3), the scores of work productivity and life satisfaction also significantly decreased (SAS scores from 2.8 to 1.3, SDS scores from 18.2 to 2.8, and LS scores from 14.9 to 7.1, respectively). Following the acute treatment period, work productivity and life satisfaction further continuously improved till the end of the maintenance treatment phase. CONCLUSION: Parallel to the improvement of the depressive symptoms, the number of absence days decreased, the work performance, free time activity and family functioning significantly improved. A significant correlation was found between the improvement of depressive symptoms and the positive changes in work production and life satisfaction.


Subject(s)
Antidepressive Agents, Second-Generation/therapeutic use , Depressive Disorder/psychology , Efficiency/drug effects , Paroxetine/therapeutic use , Personal Satisfaction , Selective Serotonin Reuptake Inhibitors/therapeutic use , Adult , Aged , Comorbidity , Depressive Disorder/drug therapy , Employment , Female , Follow-Up Studies , Humans , Male , Middle Aged , Product Surveillance, Postmarketing , Prospective Studies , Quality of Life , Severity of Illness Index , Surveys and Questionnaires , Treatment Outcome , Work
20.
Regul Pept ; 123(1-3): 51-9, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15518893

ABSTRACT

Pituitary adenylate cyclase activating polypeptide (PACAP) exerts neuroprotective effects in various in vitro and in vivo models of cerebral pathologies. It has been shown that PACAP protects neurons in rat models of both global and focal ischemia. In the present study, we investigated factors that may play a role in the neuroprotective effects of PACAP. PACAP strongly reduced the anisomycin-induced apoptosis of PC12 cells, which was abolished in a PKA-deficient PC12 cell line (A126). This effect was also observed in vivo, in permanent occlusion of the middle cerebral artery, where the number of TUNEL-positive neurons was significantly reduced in the ischemic core of PACAP-treated animals. Our results show that PACAP has a minor antioxidant effect in a non-cellular in vitro system, and has considerable antioxidant effects in an in vitro red blood cell filtration model. PACAP had no effect on platelet aggregation induced by collagen, ADP or epinephrine. Our results demonstrate that the effects of PACAP on delayed neuronal death may play a significant role in the reduction of the infarct size in vivo, but the antioxidant effect could only be observed at concentrations higher than that used in the model of focal ischemia.


Subject(s)
Nerve Growth Factors/pharmacology , Neurons/drug effects , Neuropeptides/pharmacology , Neurotransmitter Agents/pharmacology , Platelet Aggregation/drug effects , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/prevention & control , Erythrocyte Deformability/drug effects , Humans , In Vitro Techniques , Leukocytes/drug effects , Leukocytes/metabolism , Male , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/pharmacology , PC12 Cells , Pituitary Adenylate Cyclase-Activating Polypeptide , Rats , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...