scispace - formally typeset
Search or ask a question

Showing papers in "Current Medicinal Chemistry in 2014"


Journal ArticleDOI
Yixi Xie, Weijie Yang, Fen Tang, Xiaoqing Chen, Licheng Ren1 
TL;DR: It is concluded that hydroxyls at special sites on the aromatic rings of flavonoids improve the activity, however, the methylation of the active Hydroxyl groups generally decreases the activity.
Abstract: Flavonoids are well known as antibacterial agents against a wide range of pathogenic microorganism. With increasing prevalence of untreatable infections induced by antibiotic resistance bacteria, flavonoids have attracted much interest because of the potential to be substitutes for antibiotics. In this review, the structure-relationship of flavonoids as antibacterial agents is summarized, and the recent advancements on the antibacterial mechanisms of flavonoids are also discussed. It is concluded that hydroxyls at special sites on the aromatic rings of flavonoids improve the activity. However, the methylation of the active hydroxyl groups generally decreases the activity. Besides, the lipopholicity of the ring A is vital for the activity of chalcones. The hydrophobic substituents such as prenyl groups, alkylamino chains, alkyl chains, and nitrogen or oxygen containing heterocyclic moieties usually enhance the activity for all the flavonoids. The proposed antibacterial mechanisms of flavonoids are as follows: inhibition of nucleic acid synthesis, inhibition of cytoplasmic membrane function, inhibition of energy metabolism, inhibition of the attachment and biofilm formation, inhibition of the porin on the cell membrane, alteration of the membrane permeability, and attenuation of the pathogenicity.

428 citations


Journal ArticleDOI
TL;DR: The benefits of dietary polyphenols for type 2 diabetes can be summarized as: protection of pancreatic β-cells against glucose toxicity, anti-inflammatory and antioxidant effects, inhibition of α-amylases or α- glucosidases and thus decrease of starch digestion, and inhibition of advanced glycation end products formation.
Abstract: Significant evidence suggests that polyphenol-rich diets have the ability to protect against diabetes. Since several previous reviews focused on the nutrition and health effects including type 2 diabetes of polyphenols in 2007-2008, a number of related original publications have been pulished in this field. This review summarizes important advances related to influence of dietary polyphenols and polyphenol-rich diets on preventing and managing type 2 diabetes, as well as diabetes-mediated changes in bioactivities of dietary polyphenols. It appears that anthocyanins or anthocyanin-rich food intake is related to the risk of type 2 diabetes, but there is no association for other polyphenol subclasses. It is discussed that procyanidins are more active when administered individually than when mixed with food. The benefits of dietary polyphenols for type 2 diabetes can be summarized as: protection of pancreatic β-cells against glucose toxicity, anti-inflammatory and antioxidant effects, inhibition of α-amylases or α- glucosidases and thus decrease of starch digestion, and inhibition of advanced glycation end products formation. Moreover, type 2 diabetes also significantly influences the benefits of dietary polyphenols, although there are very limited studies have been conducted so far. How type 2 diabetes impacts the pharmacology of dietary polyphenols is not well understood. Comprehension of type 2 diabetes-mediated changes in pharmacokinetics and bioactivity of dietary polyphenols might lead to improve the benefits of these phytochemicals and subsequent clinical outcomes for type 2 diabetics.

286 citations


Journal ArticleDOI
TL;DR: The majority of the recently reported structures are acetylcholinesterase inhibitors, often endowed with some additional properties giving hope for not only symptomatic but also causal treatment of the disease.
Abstract: Alzheimer's disease (AD) is a complex and progressive neurodegenerative disorder. The available therapy is limited to the symptomatic treatment and its efficacy remains unsatisfactory. In view of the prevalence and expected increase in the incidence of AD, the development of an effective therapy is crucial for public health. Due to the multifactorial aetiology of this disease, the multi-target-directed ligand (MTDL) approach is a promising method in search for new drugs for AD. This review updates information on the development of multifunctional potential anti-AD agents published within the last three years. The majority of the recently reported structures are acetylcholinesterase inhibitors, often endowed with some additional properties. These properties enrich the pharmacological profile of the compounds giving hope for not only symptomatic but also causal treatment of the disease. Among these advantageous properties, the most often reported are an amyloid-β antiaggregation activity, inhibition of β-secretase and monoamine oxidase, an antioxidant and metal chelating activity, NOreleasing ability and interaction with cannabinoid, NMDA or histamine H3 receptors. The majority of novel molecules possess heterodimeric structures, able to interact with multiple targets by combining different pharmacophores, original or derived from natural products or existing therapeutics (tacrine, donepezil, galantamine, memantine). Among the described compounds, several seem to be promising drug candidates, while others may serve as a valuable inspiration in the search for new effective therapies for AD.

251 citations


Journal ArticleDOI
TL;DR: While the enzymes involved in oxidative stress have been identified, the temporal course of this imbalance following TBI has yet to be determined, which would explain why most antioxidant strategies developed to treat patients with TBI have failed.
Abstract: Traumatic brain injury (TBI) is a major healthcare concern, constituting a major cause of death and disability throughout the world. Among the factors leading to TBI outcome are biochemical cascades which occur in response to primary and secondary injury. These mechanisms generate oxidative stress, an imbalance between oxidant and antioxidant agents that can result in neural dysfunction and death. After TBI, an assembly of oxidative stress markers (carbonylated proteins, lipid peroxides, reactive oxygen and reactive nitrogen species) are produced in the brain, while antioxidant defense enzymes decrease (GSH, ratio GSH/GSSG, GPx, GR, GST, G-6PD, SOD, CAT). This imbalance is directly related to the pathogenesis of TBI. Therefore, the development of antioxidant strategies is of primary interest in ongoing efforts to optimize brain injury treatment. The success of any drug intervention strategy relies, in part, on knowledge of the optimal dosage and therapeutic window for its administration. But while the enzymes involved in oxidative stress have been identified, the temporal course of this imbalance following TBI has yet to be determined. This would explain why most antioxidant strategies developed to treat patients with TBI have failed.

209 citations


Journal ArticleDOI
TL;DR: A better understanding of the roles of the different immune cells and their temporal profile of damage versus repair will help to clarify more effective modulation of inflammation post stroke.
Abstract: Stroke is a frequent cause of long-term disability and death worldwide. Ischemic stroke is more commonly encountered compared to hemorrhagic stroke, and leads to tissue death by ischemia due to occlusion of a cerebral artery. Inflammation is known to result as a result of ischemic injury, long thought to be involved in initiating the recovery and repair process. However, work over the past few decades indicates that aspects of this inflammatory response may in fact be detrimental to stroke outcome. Acutely, inflammation appears to have a detrimental effect, and anti-inflammatory treatments have been been studied as a potential therapeutic target. Chronically, reports suggest that post-ischemic inflammation is also essential for the tissue repairing and remodeling. The majority of the work in this area has centered around innate immune mechanisms, which will be the focus of this review. This review describes the different key players in neuroinflammation and their possible detrimental and protective effects in stroke. A better understanding of the roles of the different immune cells and their temporal profile of damage versus repair will help to clarify more effective modulation of inflammation post stroke.

202 citations


Journal ArticleDOI
TL;DR: A summary of what is known on the toxicology related to the specificity of nanoparticles, both as technological tools or ambient pollutants is provided to highlight their potential hazard and to provide a balanced update on all the important questions and directions that should be focused in the near future.
Abstract: Nowadays more than thousands of different nanoparticles are known, though no well-defined guidelines to evaluate their potential toxicity and to control their exposure are fully provided. The way of entry of nanoparticles together with their specificities such as chemistry, chemical composition, size, shape or morphology, surface charge and area can influence their biological activities and effects. A specific property may give rise to either a safe particle or to a dangerous one. The small size allows nanoparticles to enter the body by crossing several barriers, to pass into the blood stream and lymphatic system from where they can reach organs and tissues and strictly interact with biological structures, thus damaging their normal functions in different ways. This review provides a summary of what is known on the toxicology related to the specificity of nanoparticles, both as technological tools or ambient pollutants. The aim is to highlight their potential hazard and to provide a balanced update on all the important questions and directions that should be focused in the near future.

188 citations


Journal ArticleDOI
TL;DR: The current knowledge on mechanisms supposed to be involved in epileptogenesis, as well as on the possible innovative treatments that may lead to a preventive approach to TLE are addressed.
Abstract: Temporal lobe epilepsy (TLE) is frequently associated with hippocampal sclerosis, possibly caused by a primary brain injury that occurred a long time before the appearance of neurological symptoms. This type of epilepsy is characterized by refractoriness to drug treatment, so to require surgical resection of mesial temporal regions involved in seizure onset. Even this last therapeutic approach may fail in giving relief to patients. Although prevention of hippocampal damage and epileptogenesis after a primary event could be a key innovative approach to TLE, the lack of clear data on the pathophysiological mechanisms leading to TLE does not allow any rational therapy. Here we address the current knowledge on mechanisms supposed to be involved in epileptogenesis, as well as on the possible innovative treatments that may lead to a preventive approach. Besides loss of principal neurons and of specific interneurons, network rearrangement caused by axonal sprouting and neurogenesis are well known phenomena that are integrated by changes in receptor and channel functioning and modifications in other cellular components. In particular, a growing body of evidence from the study of animal models suggests that disruption of vascular and astrocytic components of the blood-brain barrier takes place in injured brain regions such as the hippocampus and piriform cortex. These events may be counteracted by drugs able to prevent damage to the vascular component, as in the case of the growth hormone secretagogue ghrelin and its analogues. A thoroughly investigation on these new pharmacological tools may lead to design effective preventive therapies.

177 citations


Journal ArticleDOI
TL;DR: The role of theNFκB signaling pathways in the development and progression of breast cancer and the validity of NFκB as a potential target for breast cancer prevention and therapy are focused on.
Abstract: The activation of nuclear factor-kappaB (NFκB), a proinflammatory transcription factor, is a commonly observed phenomenon in breast cancer. It facilitates the development of a hormone-independent, invasive, high-grade, and late-stage tumor phenotype. Moreover, the commonly used cancer chemotherapy and radiotherapy approaches activate NFκB, leading to the development of invasive breast cancers that show resistance to chemotherapy, radiotherapy, and endocrine therapy. Inhibition of NFκB results in an increase in the sensitivity of cancer cells to the apoptotic effects of chemotherapeutic agents and radiation and restoring hormone sensitivity, which is correlated with increased disease-free survival in patients with breast cancer. In this review article, we focus on the role of the NFκB signaling pathways in the development and progression of breast cancer and the validity of NFκB as a potential target for breast cancer prevention and therapy. We also discuss the recent findings that NFκB may have tumor suppressing activity in certain cancer types. Finally, this review also covers the state-of-the-art development of NFκB inhibitors for cancer therapy and prevention, the challenges in targeting validation, and pharmacology and toxicology evaluations of these agents from the bench to the bedside.

154 citations


Journal ArticleDOI
TL;DR: The mechanisms of HHcy-induced endothelial dysfunction and the metabolism and physiological functions of H2S as a protective agent are summarized.
Abstract: High level of homocysteine (hyperhomocysteinemia, HHcy) is associated with increased risk for vascular disease. Evidence for this emerges from epidemiological studies which show that HHcy is associated with premature peripheral, coronary artery and cerebrovascular disease independent of other risk factors. Possible mechanisms by which homocysteine causes vascular injury include endothelial injury, DNA dysfunction, proliferation of smooth muscle cells, increased oxidative stress, reduced activity of glutathione peroxidase and promoting inflammation. HHcy has been shown to cause direct damage to endothelial cells both in vitro and in vivo. Clinically, this manifests as impaired flow-mediated vasodilation and is mainly due to a reduction in nitric oxide synthesis and bioavailability. The effect of impaired nitric oxide release can in turn trigger and potentiate atherothrombogenesis and oxidative stress. Endothelial damage is a crucial aspect of atherosclerosis and precedes overt manifestation of disease. In addition, endothelial dysfunction is also associated with hypertension, diabetes, ischemia reperfusion injury and neurodegenerative diseases. Homocysteine is a precursor of hydrogen sulfide (H2S) which is formed by transulfuration process catalyzed by the enzymes, cystathionine β-synthase and cystathionine γ-lyase. H2S is a gasotransmitter that has emerged recently as a novel mediator in cardiovascular homeostasis. As a potent vasodilator, it plays several roles which include regulation of vessel diameter, protection of endothelium from redox stress, ischemia reperfusion injury and chronic inflammation. However, the precise mechanism by which it mediates these beneficial effects is complex and still remains unclear. Current evidence indicates H2S modulates cellular functions by a variety of intracellular signaling processes. In this review, we summarize the mechanisms of HHcy-induced endothelial dysfunction and the metabolism and physiological functions of H2S as a protective agent.

151 citations


Journal ArticleDOI
TL;DR: This review is focused on antibacterial (against Gram (-) and Gram (+) bacteria) and antifungal peptides, discussing action mode of AMPs, and recent advances in the study of the molecular basis of their anti-microbial activity.
Abstract: In the last decades, the indiscriminate use of conventional antibiotics has generated high rates of microbial resistance. This situation has increased the need for obtaining new antimicrobial compounds against infectious diseases. Among these, antimicrobial peptides (AMPs) constitute a promising alternative as therapeutic agents against various pathogenic microbes. These therapeutic agents can be isolated from different organisms, being widespread in nature and synthesized by microorganisms, plants and animals (both invertebrates and vertebrates). Additionally, AMPs are usually produced by a non-specific innate immune response. These peptides are involved in the inhibition of cell growth and in the killing of several microorganisms, such as bacteria, fungi, enveloped viruses, protozoans and other parasites. They have many interesting properties as potential antibiotics, such as relatively small sizes (below 25-30 kDa), amphipathic structures, cationic nature, and offer low probability for the generation of microbial resistance. In recent years, many novel AMPs, with very promising therapeutic properties, have been discovered. These peptides have been the base for the production of chemical analogs, which have been designed, chemically synthesized and tested in vitro for their antimicrobial activity. This review is focused on antibacterial (against Gram (-) and Gram (+) bacteria) and antifungal peptides, discussing action mode of AMPs, and recent advances in the study of the molecular basis of their anti-microbial activity. Finally, we emphasize on their current pharmacological development, future directions and applications of AMPs as promising antibiotics of therapeutic use for microbial infections.

146 citations


Journal ArticleDOI
TL;DR: It is possible to develop pharmacologically relevant cytotoxic agents by specifically inhibiting CDK2 activity with lesser toxicity than traditional chemotherapeutic agents by targeting a binding pocket other than ATP.
Abstract: Cyclin-dependent kinase-2 (CDK2) is a member of protein kinase family. It plays an important role in regulating various events of eukaryotic cell division cycle. Accumulated evidences indicated that over expression of CDK2 should cause the abnormal regulation of cell-cycle, which would be directly associated with hyperproliferation in cancer cells. Therefore, CDK2 was regarded as a potentially therapeutic target for cancer therapy. Knowledge of crystallography and availability of X-ray crystal structure of CDK2 have enabled us to understand the mode of CDK2 inhibition, which facilitated the development of numerous CDK2 inhibitors. Some of the CDK2 inhibitors were investigated clinically for their potential as anti-cancer agents. In this review, we present the structure, functions and activation of CDK2 by cyclin binding with special focus on recent advances in the development of different classes of CDK2 inhibitors. We also summarize different strategies to achieve subtype specificity either by targeting a binding pocket other than ATP, i.e. allosteric ligand binding site or by natural protein inhibitors capable to disrupt CDK2-cyclin complexes. It is possible to develop pharmacologically relevant cytotoxic agents by specifically inhibiting CDK2 activity with lesser toxicity than traditional chemotherapeutic agents.

Journal ArticleDOI
TL;DR: This review focuses on the recent developments in antimicrobial surface coatings with respect to biomaterial implants and devices, and the use of polymer chains, or hydrogels is preferred for anti-adhesive strategies.
Abstract: Bacterial adhesion and subsequent biofilm formation on material surfaces represent a serious problem in society from both an economical and health perspective. Surface coating approaches to prevent bacterial adhesion and biofilm formation are of increased importance due to the increasing prevalence of antibiotic resistant bacterial strains. Effective antimicrobial surface coatings can be based on an anti-adhesive principle that prevents bacteria to adhere, or on bactericidal strategies, killing organisms either before or after contact is made with the surface. Many strategies, however, implement a multifunctional approach that incorporates both of these mechanisms. For anti-adhesive strategies, the use of polymer chains, or hydrogels is preferred, although recently a new class of super-hydrophobic surfaces has been described which demonstrate improved anti-adhesive activity. In addition, bacterial killing can be achieved using antimicrobial peptides, antibiotics, chitosan or enzymes directly bound, tethered through spacer-molecules or encased in biodegradable matrices, nanoparticles and quaternary ammonium compounds. Notwithstanding the ubiquitous nature of the problem of microbial colonization of material surfaces, this review focuses on the recent developments in antimicrobial surface coatings with respect to biomaterial implants and devices. In this biomedical arena, to rank the different coating strategies in order of increasing efficacy is impossible, since this depends on the clinical application aimed for and whether expectations are short- or long term. Considering that the era of antibiotics to control infectious biofilms will eventually come to an end, the future for biofilm control on biomaterial implants and devices is likely with surface-associated modifications that are non-antibiotic related.

Journal ArticleDOI
TL;DR: An overview of excitotoxicity in ALS is provided, focusing on the events that contribute to excess glutamate, how the excess might damage nerve cells, and how this information is being harnessed in the development of potential new neuroprotective agents.
Abstract: Amyotrophic lateral sclerosis (ALS) is an age-related neurodegenerative disorder that is believed to have complex genetic and environmental influences in the pathogenesis, but etiologies are unidentified for most patients. Until the major causes are better defined, drug development is directed at downstream pathophysiological mechanisms, themselves incompletely understood. For nearly 30 years, glutamate-induced excitotoxicity has lain at the core of theories behind the spiraling events, including mitochondrial dysfunction, oxidative stress, and protein aggregation, that lead to neurodegenerative cell death. One drug, riluzole, which possesses anti-glutamatergic properties, is approved as neuroprotective for ALS. Following the achievement of the riluzole trials, numerous other agents with similar mechanisms have been tested without success. This article provides an overview of excitotoxicity in ALS, focusing on the events that contribute to excess glutamate, how the excess might damage nerve cells, and how this information is being harnessed in the development of potential new neuroprotective agents. The work highlights clinical trials of drugs that have targeted the glutamate system, comments on the potential role of glutamate as a biomarker and concludes with a section on future directions for the field. As research uncovers elusive etiologies and brings clarity to pathophysiological mechanisms, the success of new interventions will increasingly depend on the design of agents that target particular mechanisms for specific individuals. The heady future of personalized drug regimens for ALS rests with medicinal chemists, the scientists whose ideas and work produce these designer drugs.

Journal ArticleDOI
TL;DR: The mitochondrial function deteriorates with aging, and the mitochondrial damage correlates with increased intracellular production of oxidants and pro-oxidants, which contributes to the dysfunction of the blood-brain barrier (BBB) and damage to the brain parenchymal cells.
Abstract: Mitochondrial dysfunction plausibly underlies the aging-associated brain degeneration Mitochondria play a pivotal role in cellular bioenergetics and cell-survival Oxidative stress consequent to chronic hypoperfusion induces mitochondrial damage, which is implicated as the primary cause of cerebrovascular accidents (CVA) mediated Alzheimer's disease (AD) The mitochondrial function deteriorates with aging, and the mitochondrial damage correlates with increased intracellular production of oxidants and pro-oxidants The prolonged oxidative stress and the resultant hypoperfusion in the brain tissues stimulate the expression of nitric oxide synthase (NOS) enzymes, which further drives the formation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) The ROS and RNS collectively contributes to the dysfunction of the blood-brain barrier (BBB) and damage to the brain parenchymal cells Delineating the molecular mechanisms of these processes may provide clues for the novel therapeutic targets for CVA and AD patients

Journal ArticleDOI
TL;DR: The aim of this review is to depict the physical and biochemical properties of cigarette smoke and elucidate the main pathophysiological mechanisms of cigarette-induced atherosclerosis and overview the new therapeutic approaches for smoking cessation and augmentation of cardiovascular health.
Abstract: It has been clear that at least 1 billion adults worldwide are smokers and at least 700 million children are passive smokers at home. Smoking exerts a detrimental effect to many organ systems and is responsible for illnesses such as lung cancer, pneumonia, chronic obstructive pulmonary disease, cancer of head and neck, cancer of the urinary and gastrointestinal tract, periodontal disease, cataract and arthritis. Additionally, smoking is an important modifiable risk factor for the development of cardiovascular disease such as coronary artery disease, stable angina, acute coronary syndromes, sudden death, stroke, peripheral vascular disease, congestive heart failure, erectile dysfunction and aortic aneurysms via initiation and progression of atherosclerosis. A variety of studies has proved that cigarette smoking induces oxidative stress, vascular inflammation, platelet coagulation, vascular dysfunction and impairs serum lipid pro-file in both current and chronic smokers, active and passive smokers and results in detrimental effects on the cardiovascular system. The aim of this review is to depict the physical and biochemical properties of cigarette smoke and, furthermore, elucidate the main pathophysiological mechanisms of cigarette-induced atherosclerosis and overview the new therapeutic approaches for smoking cessation and augmentation of cardiovascular health.

Journal ArticleDOI
TL;DR: The major preclinical and clinical evidences of MDM2 inhibitors for human cancer treatment are summarized and suggestions to further improve efficacy and safety of this interesting class of cancer therapeutics are made.
Abstract: Inactivation of the tumor suppressor p53 and/or overexpression of the oncogene MDM2 frequently occur in human cancers, and are associated with poor prognosis, advanced forms of the disease, and chemoresistance. MDM2, the major negative regulator of p53, induces p53 degradation and inactivates its tumor suppressing activity. In turn, p53 regulates MDM2 expression. This MDM2-p53 negative feedback loop has been widely studied and presents an attractive target for cancer therapy, with a few of the inhibitors of this interaction already having advanced into clinical trials. Additionally, there is an increasing interest in understanding MDM2's p53-independent activities in carcinogenesis and cancer progression, which may also have implications for cancer therapy. This review aims to highlight the various roles that the MDM2-p53 interaction plays in cancer, the p53 independent oncogenic activities of MDM2 and the various strategies that may be used to target MDM2 and the MDM2-p53 interaction. We will summarize the major preclinical and clinical evidences of MDM2 inhibitors for human cancer treatment and make suggestions to further improve efficacy and safety of this interesting class of cancer therapeutics.

Journal ArticleDOI
TL;DR: The protective role of PPARα agonists in neurodegenerative diseases and in neuropsychiatric disorders makes manipulation of this pathway highly attractive as therapeutic strategy for neuropathological conditions.
Abstract: Signalling lipids are known to control a wide array of cellular processes, including cell proliferation, apoptosis, migration, and energy metabolism. Fatty acids and their derivatives, eicosanoids, phosphoinositides, sphingolipids, some cannabinoid-like molecules bind and activate nuclear receptors, including peroxisome proliferator-activated receptors (PPARs). This subfamily of transcription factors comprises three isotypes - PPARα (NR1C1), PPAR β/δ (NR1C2), PPARγ (NR1C3) - which bind to specific DNA response elements, as heterodimers with retinoid X receptors. PPAR activity is modulated by post-translational modifications and cofactors, towards which they show differential affinity. The three PPARs mutually interact, being integrated in a complex system, leading to the concept of a "PPAR triad". Nevertheless, the isotypes also show distinct actions on cellular physiology and partially different tissue, ligand and target gene specificities. In the brain, while the functions of PPARγ and its ligands are being thoroughly investigated, the actual and potential roles of PPARα and β/δ are far from being clarified. PPARα appears especially intriguing, since it is selectively expressed in certain brain areas and neuronal/glial populations, and modulates antioxidant responses, neurotransmission, neuroinflammation, neurogenesis, and glial cell proliferation/differentiation. This receptor and its endogenous ligands, including oleoylethanoloamide (OEA) and palmitoylethanolamide (PEA), are involved in physiological and pathological responses, such as satiety, memory consolidation, and modulation of pain perception. The protective role of PPARα agonists in neurodegenerative diseases and in neuropsychiatric disorders makes manipulation of this pathway highly attractive as therapeutic strategy for neuropathological conditions. In this review, we focus on the pleiotropic functions of PPARα and its lipid ligands in the nervous tissue, devoting special attention to neuroprotection.

Journal ArticleDOI
TL;DR: This review describes the classification of the nearly 60 pyoverdines identified so far, their structural and chemical properties and their (bio)synthesis.
Abstract: Pyoverdine is the generic name given to a vast family of fluorescent green-yellowish pigments produced by Pseudomonas species. Pseudomonas aeruginosa is an opportunistic pathogen, particularly infecting humans with compromised natural defenses. These infections result in significantly higher morbidity, longer hospitalization, increased mortality rates and excess health care costs. P. aeruginosa is very difficult to eradicate because of an intrinsic coupled with an adaptive resistance to a wide variety of classical antibiotics. When subjected to iron starvation conditions, Pseudomonas bacteria synthesize pyoverdines, their primary siderophores, to acquire iron from the extracellular medium. These molecules are not only powerful iron(III) scavengers but efficient iron(III) transporters as well. Three distinct structural parts constitute pyoverdines, i.e. (i) the fluorescent chromophore, deriving from a dihydroxyquinoline, attached via its carbonyl group to (ii) a type-specific peptide composed of 6 to 14 amino acids and (iii) a small side chain corresponding to a carboxylic acid derivative. Their chemical structure show three bidentate chelating sites including a catechol and two hydroxamates, leading to an octahedral geometry when complexed to iron(III). While the chromophore group is common to all pyoverdines, their peptide moiety differs among strains and species by the number, length, composition and configuration of amino acids. Following chelation with iron(III), the newly formed pyoverdine-Fe complex is recognized by a specific outer membrane transporter, namely FpvA, and reenters the cell where the iron is released from the pyoverdine into the periplasm for further incorporation into bacterial proteins. The remaining apo-pyoverdine is then recycled and secreted back to the extracellular medium by efflux pumps. Besides, the role of pyoverdines in P. aeruginosa is not only limited to scavenge iron from the bacterial environment. Indeed, these siderophores act as signal molecules for the production of acute virulence factors and are involved in biofilm formation as well. The ongoing expanding pathogenicity of P. aeruginosa has become a major public health problem, and finding alternative strategies to classical antibiotics is urgently needed. Pyoverdines along with the iron pathway recently gained interest among academical researchers as potential new approaches to “fight” the bacteria. This review describes the classification of the nearly 60 pyoverdines identified so far, their structural and chemical properties and their (bio)synthesis. The different mechanisms underlying the steps of a pyoverdine’s life in Pseudomonas are detailed as well: the affinity by which a pyoverdine chelates iron(III), the description of the interactions inducing the siderophore-receptor recognition, the specific transport of the pyoverdine-Fe(III) complex. As pyoverdine production and severe infections are linked, we will also report on situations where pyoverdines are considered as being P. aeruginosa Achilles heel: the propensity of FpvA to transport exo-pyoverdines, organic synthesis of pyoverdines and analogs, grafting of antibiotics on pyoverdines in a Trojan Horse strategy.

Journal ArticleDOI
TL;DR: Using structure-based drug design processes, it is estimated that it will be possible to achieve the desired selectivity for inhibiting preferentially the bacterial but not the host CA isoforms.
Abstract: The possibility to develop new antibacterial agents raised much interest recently. The main classes of antibiotics clinically used nowadays act towards the inhibition of four classical targets: a) cell wall biosynthesis; b) protein biosynthesis; c) DNA and RNA biosynthesis; d) folate biosynthesis. Recently, carbonic anhydrases (CAs, EC 4.2.1.1) started to be investigated in detail in pathogenic bacteria, in the search for antibiotics with a novel mechanism of action, since it has been demonstrated that in many bacteria, CAs are essential for the life cycle of the organism and that their inhibition leads to growth impairment or growth defects of the pathogen. CAs catalyze a simple but physiologically relevant reaction in all life kingdoms, carbon dioxide hydration to bicarbonate and protons. Several classes of CA inhibitors (CAIs) are known to date: the metal complexing anions and the unsubstituted sulfonamides, which bind to the Zn(II) ion of the enzyme either by substituting the non-protein zinc ligand or add to the metal coordination sphere, generating trigonal- bipyramidal species are the classical, most frequently investigated ones. In many cases effective inhibitors were detected, some of which also inhibited the bacterial growth in vivo. However, very few of the detected inhibitors were also selective for the bacterial over the human, off target isoforms such as hCA II. Using structure-based drug design processes, we estimate that it will be possible to achieve the desired selectivity for inhibiting preferentially the bacterial but not the host CA isoforms.

Journal ArticleDOI
TL;DR: An overview of the current understanding of the role of Hif-1 in therapeutic resistance is provided and the ongoing effort to develop HIF-1 inhibitors as an anti-cancer strategy is discussed.
Abstract: Activation of hypoxia-inducible factor 1 (HIF-1) signaling is observed in a broad range of human cancers due to tumor hypoxia and epigenetic mechanisms. HIF-1 activation leads to the transcription of a plethora of target genes that promote physiological changes associated with therapeutic resistance, including the inhibition of apoptosis and senescence and the activation of drug efflux and cellular metabolism. As a result, targeting HIF-1 represents an attractive strategy to enhance the efficacy of current therapies as well as reduce resistance to chemotherapy in tumors. Approaches to inhibit HIF-1 signaling have primarily focused on reducing HIF-1α protein levels, by inducing its degradation or inhibiting its transcription, inhibiting HIF-1-mediated transcription, or disrupting the formation of the HIF-1 transcription factor complex. To date, multiple preclinical and clinical agents have been identified that effectively inhibit HIF-1 activity through various mechanisms, likely accounting for a portion of their anti-tumor efficacy. This review aims to provide an overview of our current understanding of the role of HIF-1 in therapeutic resistance and discuss the ongoing effort to develop HIF-1 inhibitors as an anti-cancer strategy.

Journal ArticleDOI
TL;DR: The review leads to a novel framework for successful therapeutic modification of several cardinal pathophysiological processes leading to adverse cardiovascular outcome, including TRP breakdown and related complex mechanisms on the prognosis and individual course of CVD.
Abstract: Atherosclerosis (AS), a major pathologic consequence of obesity, is the main etiological factor of cardiovascular disease (CVD), which is the most common cause of death in the western world. A systemic chronic low grade immune- mediated inflammation (scLGI) is substantially implicated in AS and its consequences. In particular, proinflammatory cytokines play a major role, with Th1-type cytokine interferon-γ (IFN-γ) being a key mediator. Among various other molecular and cellular effects, IFN-γ activates the enzyme indoleamine 2,3-dioxygenase (IDO) in monocyte-derived macrophages, dendritic, and other cells, which, in turn, decreases serum levels of the essential amino acid tryptophan (TRP). Thus, people with CVD often have increased serum kynurenine to tryptophan ratios (KYN/TRP), a result of an increased TRP breakdown. Importantly, increased KYN/TRP is associated with a higher likelihood of fatal cardiovascular events. A scLGI with increased production of the proinflammatory adipokine leptin, in combination with IFN-γ and interleukin-6 (IL-6), represents another central link between obesity, AS, and CVD. Leptin has also been shown to contribute to Th1-type immunity shifting, with abdominal fat being thus a direct contributor to KYN/TRP ratio. However, TRP is not only an important source for protein production but also for the generation of one of the most important neurotransmitters, 5-hydroxytryptamine (serotonin), by the tetrahydrobiopterin-dependent TRP 5-hydroxylase. In prolonged states of scLGI, availability of free serum TRP is strongly diminished, affecting serotonin synthesis, particularly in the brain. Additionally, accumulation of neurotoxic KYN metabolites such as quinolinic acid produced by microglia, can contribute to the development of depression via NMDA glutamatergic stimulation. Depression had been reported to be associated with CVD endpoints, but it most likely represents only a secondary loop connecting excess adipose tissue, scLGI and cardiovascular morbidity and mortality. Accelerated catabolism of TRP is further involved in the pathogenesis of the anemia of scLGI. The pro-inflammatory cytokine IFN-γ suppresses growth and differentiation of erythroid progenitor cells, and the depletion of TRP limits protein synthesis and thus hemoglobin production, and, through reduction in oxygen supply, may contribute to ischemic vascular disease. In this review we discuss the impact of TRP breakdown and the related complex mechanisms on the prognosis and individual course of CVD. Measurement of TRP, KYN concentrations, and calculation of the KYN/TRYP ratio will contribute to a better understanding of the interplay between inflammation, metabolic syndrome, mood disturbance, and anemia, all previously described as significant predictors of an unfavorable outcome in patients with CVD. The review leads to a novel framework for successful therapeutic modification of several cardinal pathophysiological processes leading to adverse cardiovascular outcome.

Journal ArticleDOI
TL;DR: It is important to understand the proper dose and duration of supplementation with polyphenols-rich extracts in order to guide effective therapeutic interventions in diabetic patients.
Abstract: Currently, experimental and clinical evidences showed that polyphenols-rich natural products, like nutraceuticals and food supplements, may offer unique treatment modalities in type 2 diabetes mellitus (DM), due to their biological properties. Natural products modulate the carbohydrate metabolism by various mechanisms, such as restoring beta-cells integrity and physiology, enhancing insulin releasing activity, and the glucose using. Sea buckthorn berries, red grapes, bilberries, chokeberries and popular drinks like cocoa, coffee and green tea are all rich in polyphenols and may decrease the insulin response, offer in g a natural alternative of treatment in diabetes. Therefore, researches are now focused on potential efficacies of different types of polyphenols, including flavonoids, phenolic acids, lignans, anthocyans and stilbenes. Animal and human studies showed that polyphenols modulate carbohydrate and lipid metabolism, decrease glycemia and insulin resistance, increase lipid metabolism and optimize oxidative stress and inflammatory processes. It is important to understand the proper dose and duration of supplementation with polyphenols-rich extracts in order to guide effective therapeutic interventions in diabetic patients.

Journal ArticleDOI
TL;DR: The role of different risk factors, including obesity, hypertension and hypercholesterolemia in atherosclerosis, which may ultimately lead to either cardiovascular or cerebral complication is discussed.
Abstract: Atherosclerosis is a widespread disease of the arterial system that is generated by injury to the vasculature due to hypercholesterolemia, hypertension and inflammatory diseases. In the current review, we discuss the role of different risk factors, including obesity, hypertension and hypercholesterolemia in atherosclerosis, which may ultimately lead to either cardiovascular or cerebral complication. Inflammation plays a pivotal role in conjunction with obesity, hypertension and hypercholesterolemia in the etiology of atherosclerosis. We discuss the role of inflammation with regards to reactive oxygen species (ROS) linked to the specific risk factors. The role of nitric oxide (NO) in conjunction with ROS is also important. Correlations of inflammatory cytokines and their functions in the mentioned risk factors are also discussed. The risk factors may ultimately lead to ischemic events, including transient ischemic attacks (TIAs), thrombotic stroke and myocardial infarction. Importantly, it seems as if there is a combination of pathophysiological triggers that may eventually result in atherosclerosis. Therefore, atherosclerosis is not the result of only one risk factor, but a combination of various physiological processes such as homeostasis and the inflammatory response. Ultimately, each patient's risk profile is unique and determines their immediate risk for acute thrombotic events or lethal ischemia.

Journal ArticleDOI
TL;DR: It is concluded that EGCG is a promising therapeutic to combat cardiovascular complications associated with the metabolic diseases characterized by reciprocal relationships between insulin resistance and endothelial dysfunction that include obesity, metabolic syndrome and type 2 diabetes.
Abstract: Epidemiological studies demonstrate robust correlations between green tea consumption and reduced risk of type 2 diabetes and its cardiovascular complications. However, underlying molecular, cellular, and physiological mechanisms remain incompletely understood. Health promoting actions of green tea are often attributed to epigallocatechin gallate (EGCG), the most abundant polyphenol in green tea. Insulin resistance and endothelial dysfunction play key roles in the pathogenesis of type 2 diabetes and its cardiovascular complications. Metabolic insulin resistance results from impaired insulin-mediated glucose disposal in skeletal muscle and adipose tissue, and blunted insulin-mediated suppression of hepatic glucose output that is often associated with endothelial/ vascular dysfunction. This endothelial dysfunction is itself caused, in part, by impaired insulin signaling in vascular endothelium resulting in reduced insulin-stimulated production of NO in arteries, and arterioles that regulate nutritive capillaries. In this review, we discuss the considerable body of literature supporting insulin-mimetic actions of EGCG that oppose endothelial dysfunction and ameliorate metabolic insulin resistance in skeletal muscle and liver. We conclude that EGCG is a promising therapeutic to combat cardiovascular complications associated with the metabolic diseases characterized by reciprocal relationships between insulin resistance and endothelial dysfunction that include obesity, metabolic syndrome and type 2 diabetes. There is a strong rationale for well-powered randomized placebo controlled intervention trials to be carried out in insulin resistant and diabetic populations.

Journal ArticleDOI
TL;DR: Hydrogen sulfide (H2S) may prime endothelial cells (ECs) toward angiogenesis and contribute to wound healing, besides to its well-known ability to relax vascular smooth muscle cells (VSMCs), and thereby reducing blood pressure.
Abstract: The endothelium is a cellular monolayer that lines the inner surface of blood vessels and plays a central role in the maintenance of cardiovascular homeostasis by controlling platelet aggregation, vascular tone, blood fluidity and fibrinolysis, adhesion and transmigration of inflammatory cells, and angiogenesis. Endothelial dysfunctions are associated with various cardiovascular diseases, including atherosclerosis, hypertension, myocardial infarction, and cardiovascular complications of diabetes. Numerous studies have established the anti-inflammatory, anti-apoptotic, and anti-oxidant effects of hydrogen sulfide (H2S), the latest member to join the gasotransmitter family along with nitric oxide and carbon monoxide, on vascular endothelium. In addition, H2S may prime endothelial cells (ECs) toward angiogenesis and contribute to wound healing, besides to its well-known ability to relax vascular smooth muscle cells (VSMCs), and thereby reducing blood pressure. Finally, H2S may inhibit VSMC proliferation and platelet aggregation. Consistently, a deficit in H2S homeostasis is involved in the pathogenesis of atherosclerosis and of hyperglycaemic endothelial injury. Therefore, the application of H2S-releasing drugs or using gene therapy to increase endogenous H2S level may help restore endothelial function and antagonize the progression of cardiovascular diseases. The present article reviews recent studies on the role of H2S in endothelial homeostasis, under both physiological and pathological conditions, and its putative therapeutic applications.

Journal ArticleDOI
TL;DR: This review has discussed the progress made so far in the identification of tumor homing peptides, and their applications in cancer therapeutics, diagnosis, and theranostics.
Abstract: Cancer is one of the leading causes of mortality worldwide, with more than 10 million new cases each year. Despite the presence of several anticancer agents, cancer treatment is still not very effective. Main reasons behind this high mortality rate are the lack of screening tests for early diagnosis, and non-availability of tumor specific drug delivery system. Most of the current anticancer drugs are unable to differentiate between cancerous and normal cells, leading to systemic toxicity, and adverse side effects. In order to tackle this problem, a considerable progress has been made over the years to identify peptides, which specifically bind to the tumor cells, and tumor vasculature (tumor homing peptides). With the advances in phage display technology, and combinatorial libraries like one-bead one-compound library, several hundreds of tumor homing peptides, and their derivatives, which have potential to detect tumor in vivo, and deliver anticancer agents specifically to the tumor site, have been discovered. Currently, many tumor homing peptide-based therapies for cancer treatment and diagnosis are being tested in various phases of clinical trials. In this review, we have discussed the progress made so far in the identification of tumor homing peptides, and their applications in cancer therapeutics, diagnosis, and theranostics. In addition, a brief discussion on tumor homing peptide resource, and in silico designing of tumor homing peptides has also been provided.

Journal ArticleDOI
TL;DR: C3HOM is found to be a more potent antioxidant than melatonin or vitamin C in terms of its ability to scavenge the hydroxyl radical (HO.) and to recover oxidized horseradish peroxidase to its ground state.
Abstract: Cyclic 3-hydroxymelatonin (C3HOM) is an immediate product of melatonin's interaction with reactive oxygen species. Its presence has been detected in mice, rats and humans. In the current study, the antioxidant capacity and reducing power of this molecule have been systematically studied. C3HOM is found to be a more potent antioxidant than melatonin or vitamin C in terms of its ability to scavenge the hydroxyl radical (HO.) and to recover oxidized horseradish peroxidase to its ground state. The antioxidative mechanism of C3HOM is similar to that of the classic antioxidant, vitamin C, rather than to its precursor melatonin. C3HOM effectively prevents the oxidative degradation of cytochrome C induced by hydrogen peroxide (H2O2). It is speculated that some antioxidative activities of melatonin may be mediated by its metabolite, C3HOM. C3HOM prevents mitochondrial cytochrome C injury and, thus, it is likely to inhibit cellular apoptosis induced by the release of oxidized cytochrome C from mitochondria.

Journal ArticleDOI
TL;DR: A broad range of cutting-edgenanomedicines and nanomedicine platforms that are or can be used to improve ART delivery and the potential to clear reservoirs of viral infection are discussed.
Abstract: Human immunodeficiency virus (HIV) infection commonly results in a myriad of comorbid conditions secondary to immune deficiency. Infection also affects broad organ system function. Although current antiretroviral therapy (ART) reduces disease morbidity and mortality through effective control of peripheral viral load, restricted infection in HIV reservoirs including gut, lymphoid and central nervous system tissues, is not eliminated. What underlies these events is, in part, poor ART penetrance into each organ across tissue barriers, viral mutation and the longevity of infected cells. We posit that one means to improve these disease outcomes is through nanotechnology. To this end, this review discusses a broad range of cutting-edge nanomedicines and nanomedicine platforms that are or can be used to improve ART delivery. Discussion points include how polymer-drug conjugates, dendrimers, micelles, liposomes, solid lipid nanoparticles and polymeric nanoparticles can be harnessed to best yield cell-based delivery systems. When completely developed, such nanomedicine platforms have the potential to clear reservoirs of viral infection.

Journal ArticleDOI
TL;DR: This review will discuss recent clinical studies that have investigated the beneficial effects of α-lipoic acid on endothelial dysfunction and propose possible mechanisms involved.
Abstract: Reactive oxygen species and reactive nitrogen species promote endothelial dysfunction in old age and contribute to the development of cardiovascular diseases such as atherosclerosis, diabetes, and hypertension. α-Lipoic acid was identified as a catalytic agent for oxidative decarboxylation of pyruvate and α-ketoglutarate in 1951, and it has been studied intensively by chemists, biologists, and clinicians who have been interested in its role in energetic metabolism and protection from reactive oxygen species-induced mitochondrial dysfunction. Consequently, many biological effects of α-lipoic acid supplementation can be attributed to the potent antioxidant properties of α-lipoic acid and dihydro α-lipoic acid. The reducing environments inside the cell help to protect from oxidative damage and the reduction-oxidation status of α-lipoic acid is dependent upon the degree to which the cellular components are found in the oxidized state. Although healthy young humans can synthesize enough α-lipoic acid to scavenge reactive oxygen species and enhance endogenous antioxidants like glutathione and vitamins C and E, the level of α-lipoic acid significantly declines with age and this may lead to endothelial dysfunction. Furthermore, many studies have reported α-lipoic acid can regulate the transcription of genes associated with anti-oxidant and anti-inflammatory pathways. In this review, we will discuss recent clinical studies that have investigated the beneficial effects of α-lipoic acid on endothelial dysfunction and propose possible mechanisms involved.

Journal ArticleDOI
TL;DR: This manuscript aims to provide a summary of current research progress of graphene-based carbon materials in biosensing, drug (gene) delivery and tissue engineering, and discusses the opportunities and challenges in this rapidly growing field.
Abstract: Graphene, a truly two-dimensional (2D) and fully π-conjugated honeycomb network, exhibits many unique physical and chemical properties that are interesting in a wide range of areas. Since its discovery in 2004, graphene has been extensively studied in many different fields including nano-electronics, composite materials, energy research, catalysis and so on. Based on the fascinating action of members in the carbon family, notably zero dimensional (0D) fullerenes and one dimensional (1D) carbon nanotubes (CNTs) in biomedical areas, increasing number of reports have explored the potential of graphene for different biomedical and biotechnical applications since 2008. This manuscript aims to provide a summary of current research progress of graphene-based carbon materials in biosensing, drug (gene) delivery and tissue engineering, and discusses the opportunities and challenges in this rapidly growing field.