Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Bioorg Med Chem Lett ; 93: 129439, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37557925

ABSTRACT

Polyheterocycles are one of the most desired synthetic targets due to their numerous and valuable applications in various fields. We report the design and the parallel synthesis of novel linear oligocyclic guanidine peptidomimetics from predesigned reduced polyamides. A screening of these compounds identified active Mycobacterium tuberculosis DNA gyrase inhibitors which do not inhibit human DNA topoisomerase IIα and topoisomerase I.


Subject(s)
Mycobacterium tuberculosis , Peptidomimetics , Tuberculosis , Humans , DNA Gyrase , Peptidomimetics/pharmacology , Peptidomimetics/therapeutic use , Guanidines , Solid-Phase Synthesis Techniques , Tuberculosis/drug therapy , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/therapeutic use , Guanidine
2.
Mol Med ; 28(1): 146, 2022 12 07.
Article in English | MEDLINE | ID: mdl-36476230

ABSTRACT

The low efficiency of treatment strategies is one of the main obstacles to developing cancer inhibitors. Up to now, various classes of therapeutics have been developed to inhibit cancer progression. Peptides due to their small size and easy production compared to proteins are highly regarded in designing cancer vaccines and oncogenic pathway inhibitors. Although peptides seem to be a suitable therapeutic option, their short lifespan, instability, and low binding affinity for their target have not been widely applicable against malignant tumors. Given the peptides' disadvantages, a new class of agents called peptidomimetic has been introduced. With advances in physical chemistry and biochemistry, as well as increased knowledge about biomolecule structures, it is now possible to chemically modify peptides to develop efficient peptidomimetics. In recent years, numerous studies have been performed to the evaluation of the effectiveness of peptidomimetics in inhibiting metastasis, angiogenesis, and cancerous cell growth. Here, we offer a comprehensive review of designed peptidomimetics to diagnose and treat cancer.


Subject(s)
Neoplasms , Peptidomimetics , Humans , Peptidomimetics/pharmacology , Peptidomimetics/therapeutic use , Neoplasms/drug therapy , Peptides
3.
FASEB J ; 35(2): e21329, 2021 02.
Article in English | MEDLINE | ID: mdl-33484186

ABSTRACT

L1 syndrome is a rare developmental disorder characterized by hydrocephalus of varying severity, intellectual deficits, spasticity of the legs, and adducted thumbs. Therapy is limited to symptomatic relief. Numerous gene mutations in the L1 cell adhesion molecule (L1CAM, hereafter abbreviated L1) were identified in L1 syndrome patients, and those affecting the extracellular domain of this transmembrane type 1 glycoprotein show the most severe phenotypes. Previously analyzed rodent models of the L1 syndrome focused on L1-deficient animals or mouse mutants with abrogated cell surface expression of L1, making it difficult to test L1 function-triggering mimetic compounds with potential therapeutic value. To overcome this impasse, we generated a novel L1 syndrome mouse with a mutation of aspartic acid at position 201 in the extracellular part of L1 (p.D201N, hereafter termed L1-201) that displays a cell surface-exposed L1 accessible to the L1 mimetics. Behavioral assessment revealed an increased neurological deficit score and increased locomotor activity in male L1-201 mice carrying the mutation on the X-chromosome. Histological analyses of L1-201 mice showed features of the L1 syndrome, including enlarged ventricles and reduced size of the corpus callosum. Expression levels of L1-201 protein as well as extent of cell surface biotinylation and immunofluorescence labelling of cultured cerebellar neurons were normal. Importantly, treatment of these cultures with the L1 mimetic compounds duloxetine, crotamiton, and trimebutine rescued impaired cell migration and survival as well as neuritogenesis. Altogether, the novel L1 syndrome mouse model provides a first experimental proof-of-principle for the potential therapeutic value of L1 mimetic compounds.


Subject(s)
Genetic Diseases, X-Linked/drug therapy , Intellectual Disability/drug therapy , Neural Cell Adhesion Molecule L1/metabolism , Peptidomimetics/therapeutic use , Spastic Paraplegia, Hereditary/drug therapy , Animals , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Cerebellum/pathology , Cerebral Ventricles/metabolism , Cerebral Ventricles/pathology , Corpus Callosum/metabolism , Corpus Callosum/pathology , Duloxetine Hydrochloride/pharmacology , Duloxetine Hydrochloride/therapeutic use , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Intellectual Disability/genetics , Intellectual Disability/pathology , Locomotion , Male , Mice , Mice, Inbred C57BL , Mutation , Neural Cell Adhesion Molecule L1/genetics , Neurogenesis , Neurons/drug effects , Neurons/metabolism , Peptidomimetics/pharmacology , Phenotype , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/pathology , Toluidines/pharmacology , Toluidines/therapeutic use , Trimebutine/pharmacology , Trimebutine/therapeutic use
4.
J Pharmacol Sci ; 149(3): 124-138, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35641025

ABSTRACT

Protein-protein interactions (PPI) of co-stimulatory molecules CD2-CD58 are important in the early stage of an immune response, and increased expression of these co-stimulatory molecules is observed in the synovial region of joints in rheumatoid arthritis (RA) patients. A CD2 epitope region that binds to CD58 was grafted on to sunflower trypsin inhibitor (SFTI) template structure to inhibit CD2-CD58 PPI. The peptide was incorporated with an organic moiety dibenzofuran (DBF) in its structure. The designed peptidomimetic was studied for its ability to inhibit CD2-CD58 interactions in vitro, and its thermal and enzymatic stability was evaluated. Stability studies indicated that the grafted peptidomimetic was stable against trypsin cleavage. In vivo studies using the collagen-induced arthritis (CIA) model in mice indicated that the peptidomimetic was able to slow down the progress of arthritis, an autoimmune disease in the mice model. These studies suggest that with the grafting of organic functional groups in the stable peptide template SFTI stabilizes the peptide structure, and these peptides can be used as a template to design stable peptides for therapeutic purposes.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Helianthus , Peptidomimetics , Animals , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , CD58 Antigens/chemistry , CD58 Antigens/metabolism , Helianthus/chemistry , Helianthus/metabolism , Humans , Immunity , Immunomodulation , Mice , Peptides/pharmacology , Peptides, Cyclic/metabolism , Peptides, Cyclic/pharmacology , Peptidomimetics/pharmacology , Peptidomimetics/therapeutic use , Trypsin Inhibitors/therapeutic use
5.
Int J Mol Sci ; 23(2)2022 Jan 08.
Article in English | MEDLINE | ID: mdl-35054860

ABSTRACT

Based on the mechanism of neuropathic pain induction, a new type of bifunctional hybrid peptidomimetics was obtained for potential use in this type of pain. Hybrids consist of two types of pharmacophores that are connected by different types of linkers. The first pharmacophore is an opioid agonist, and the second pharmacophore is an antagonist of the pronociceptive system, i.e., an antagonist of the melanocortin-4 receptor. The results of tests in acute and neuropathic pain models of the obtained compounds have shown that the type of linker used to connect pharmacophores had an effect on antinociceptive activity. Peptidomimetics containing longer flexible linkers were very effective at low doses in the neuropathic pain model. To elucidate the effect of linker lengths, two hybrids showing very high activity and two hybrids with lower activity were further tested for affinity for opioid (mu, delta) and melanocortin-4 receptors. Their complexes with the target receptors were also studied by molecular modelling. Our results do not show a simple relationship between linker length and affinity for particular receptor types but suggest that activity in neuropathic pain is related to a proper balance of receptor affinity rather than maximum binding to any or all of the target receptors.


Subject(s)
Melanocortins/chemistry , Neuralgia/drug therapy , Peptidomimetics/therapeutic use , Amino Acid Sequence , Analgesics , Animals , Binding Sites , HEK293 Cells , Humans , Mice , Models, Biological , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Receptors, Opioid, mu/chemistry , Receptors, Opioid, mu/metabolism
6.
Prep Biochem Biotechnol ; 51(6): 519-529, 2021.
Article in English | MEDLINE | ID: mdl-33459157

ABSTRACT

Nowadays, proteins are frequently administered as therapeutic agents in human diseases. However, the main challenge regarding the clinical application of therapeutic proteins is short circulating plasma half-life that leads to more frequent injections for maintaining therapeutic plasma levels, increased therapy costs, immunogenic reactions, and low patient compliance. So, the development of novel strategies to enhance the pharmacokinetic profile of therapeutic proteins has attracted great attention in pharmaceuticals. So far, several techniques, each with their pros and cons, have been developed including chemical bonding to polymers, hyper glycosylation, Fc fusion, human serum albumin fusion, and recombinant PEG mimetics. These techniques mainly classify into three strategies; (i) the endosomal recycling of neonatal Fc receptor which is observed for immunoglobulins and albumin, (ii) decrease in receptor-mediated clearance, and (iii) increase in hydrodynamic radius through chemical and genetic modifications. Recently, novel PEG mimetic peptides like proline/alanine/serine repeat sequences are designed to overcome pitfalls associated with the previous technologies. Biodegradability, lack of or low immunogenicity, product homogeneity, and a simple production process, currently make these polypeptides as the preferred technology for plasma half-life extension of therapeutic proteins. In this review, challenges and pitfalls in the pharmacokinetic enhancement of therapeutic proteins using PEG-mimetic peptides will be discussed in detail.


Subject(s)
Peptides , Peptidomimetics , Recombinant Fusion Proteins , Animals , Humans , Peptides/chemistry , Peptides/pharmacokinetics , Peptides/therapeutic use , Peptidomimetics/chemistry , Peptidomimetics/pharmacokinetics , Peptidomimetics/therapeutic use , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use
7.
Molecules ; 26(22)2021 Nov 09.
Article in English | MEDLINE | ID: mdl-34833851

ABSTRACT

The vascular endothelial growth factor (VEGF) family of cytokines plays a key role in vasculogenesis, angiogenesis, and lymphangiogenesis. VEGF-A is the main member of this family, alongside placental growth factor (PlGF), VEGF-B/C/D in mammals, and VEGF-E/F in other organisms. To study the activities of these growth factors under physiological and pathological conditions, resulting in therapeutic applications in cancer and age-related macular degeneration, blocking ligands have been developed. These have mostly been large biomolecules like antibodies. Ligands with high affinities, at least in the nanomolar range, and accurate structural data from X-ray crystallography and NMR spectroscopy have been described. They constitute the main focus of this overview, which evidences similarities and differences in their binding modes. For VEGF-A ligands, and to a limited extent also for PlGF, a transition is now observed towards developing smaller ligands like nanobodies and peptides. These include unnatural amino acids and chemical modifications for designed and improved properties, such as serum stability and greater affinity. However, this review also highlights the scarcity of such small molecular entities and the striking lack of small organic molecule ligands. It also shows the gap between the rather large array of ligands targeting VEGF-A and the general absence of ligands binding other VEGF members, besides some antibodies. Future developments in these directions are expected in the upcoming years, and the study of these growth factors and their promising therapeutic applications will be welcomed.


Subject(s)
Angiogenesis Inhibitors , Macular Degeneration , Neoplasms , Neovascularization, Pathologic , Peptidomimetics , Vascular Endothelial Growth Factors , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Animals , Humans , Ligands , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Macular Degeneration/pathology , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Peptidomimetics/chemistry , Peptidomimetics/therapeutic use , Vascular Endothelial Growth Factors/chemistry , Vascular Endothelial Growth Factors/therapeutic use
8.
Org Biomol Chem ; 18(4): 583-605, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31777907

ABSTRACT

Tyrosine phosphorylation is a critical component of signal transduction for multicellular organisms, particularly for pathways that regulate cell proliferation and differentiation. While tyrosine kinase inhibitors have become FDA-approved drugs, inhibitors of the other important components of these signaling pathways have been harder to develop. Specifically, direct phosphotyrosine (pTyr) isosteres have been aggressively pursued as inhibitors of Src homology 2 (SH2) domains and protein tyrosine phosphatases (PTPs). Medicinal chemists have produced many classes of peptide and small molecule inhibitors that mimic pTyr. However, balancing affinity with selectivity and cell penetration has made this an extremely difficult space for developing successful clinical candidates. This review will provide a comprehensive picture of the field of pTyr isosteres, from early beginnings to the current state and trajectory. We will also highlight the major protein targets of these medicinal chemistry efforts, the major classes of peptide and small molecule inhibitors that have been developed, and the handful of compounds which have been tested in clinical trials.


Subject(s)
Peptidomimetics/therapeutic use , Phosphotyrosine/therapeutic use , Protein Tyrosine Phosphatases/antagonists & inhibitors , src Homology Domains/drug effects , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Cell Line, Tumor , Humans , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Phosphotyrosine/analogs & derivatives , Phosphotyrosine/pharmacology , Transcription Factors/antagonists & inhibitors
9.
Int J Mol Sci ; 21(19)2020 Oct 05.
Article in English | MEDLINE | ID: mdl-33027928

ABSTRACT

Health-care systems that develop rapidly and efficiently may increase the lifespan of humans. Nevertheless, the older population is more fragile, and is at an increased risk of disease development. A concurrently growing number of surgeries and transplantations have caused antibiotics to be used much more frequently, and for much longer periods of time, which in turn increases microbial resistance. In 1945, Fleming warned against the abuse of antibiotics in his Nobel lecture: "The time may come when penicillin can be bought by anyone in the shops. Then there is the danger that the ignorant man may easily underdose himself and by exposing his microbes to non-lethal quantities of the drug make them resistant". After 70 years, we are witnessing the fulfilment of Fleming's prophecy, as more than 700,000 people die each year due to drug-resistant diseases. Naturally occurring antimicrobial peptides protect all living matter against bacteria, and now different peptidomimetic strategies to engineer innovative antibiotics are being developed to defend humans against bacterial infections.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Peptides/therapeutic use , Peptidomimetics/therapeutic use , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/therapeutic use , Bacteria/drug effects , Bacteria/pathogenicity , Bacterial Infections/microbiology , Humans , Microbial Sensitivity Tests , Peptides/chemistry
10.
Br J Haematol ; 185(2): 219-231, 2019 04.
Article in English | MEDLINE | ID: mdl-30836448

ABSTRACT

Evasion of apoptosis has been identified as one of the essential hallmarks of cancer. Inhibitor of apoptosis proteins (IAPs) are implicated in a host of myeloid malignancies, providing the rationale for strategies aimed at neutralizing IAPs to lower the cancer cell apoptosis threshold. Modes of IAP antagonism may include down-regulating IAP expression, up-regulating endogenous pro-apoptotic proteins, such as tumour necrosis factor-α or Fas ligand, or directly antagonizing IAP activity against caspases. Direct targeting of IAPs using mimetics of the second mitochondria-derived activator of caspase (SMAC) protein has shown therapeutic promise by sensitizing the effect of chemotherapy on malignant cells. In pre-clinical studies, SMAC mimetics have demonstrated broad synergistic activity with a wide range of therapeutics, including cytotoxic chemotherapy, receptor tyrosine kinase inhibitors, agents targeting death receptors and alternative mechanisms of cell death, such as necroptosis or autophagy and immune check point blockade. SMAC mimetics represent a novel approach for further investigation in patients with high-risk, chemo-refractory blood cancers, as single agents or in thoughtfully selected combinations. In this review, we discuss the development and therapeutic rationale of small molecule SMAC mimetics, with an emphasis on agents in clinical development for myeloid malignancies.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Apoptosis Regulatory Proteins/agonists , Leukemia, Myeloid/drug therapy , Mitochondrial Proteins/agonists , Myelodysplastic Syndromes/drug therapy , Peptidomimetics/therapeutic use , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , Humans , Leukemia, Myeloid/metabolism , Molecular Targeted Therapy/methods , Myelodysplastic Syndromes/metabolism , Peptidomimetics/pharmacology
11.
Bioorg Med Chem Lett ; 29(15): 1913-1917, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31176698

ABSTRACT

The NS2B-NS3 protease is a promising target for the development of drugs against dengue virus (DENV), West Nile virus (WNV) and related flaviviruses. We report the systematic variation of the peptide backbone of the two lead compounds Bz-Arg-Lys-d-Phg-NH2 and Bz-Arg-Lys-d-Phg(OBn)-NH2. While inhibitory activity against WNV protease was generally decreased, the inhibitory potency against DENV protease could be conserved and increased in several peptidomimetics, particularly in those containing a (NMe)arginine fragment or an N-terminal α-keto amide. Methylation at the α-position of the C-terminal phenylglycine led to a 6-fold higher potency against DENV protease. Peptidomimetics with modified backbone showed increased resistance against hydrolytic attack by trypsin and α-chymotrypsin.


Subject(s)
Flavivirus/drug effects , Peptidomimetics/therapeutic use , Protease Inhibitors/therapeutic use , Humans , Molecular Structure , Peptidomimetics/pharmacology , Protease Inhibitors/pharmacology
12.
Bioorg Med Chem ; 27(4): 630-643, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30626554

ABSTRACT

It is well known that opioid analgesics produce side effects including tolerance and constipation. Since neuropeptide FF (NPFF) receptor antagonists reversed opioid-induced hyperalgesia and analgesic tolerance, the present work was performed to synthetize two branched peptidomimetics, EKR and RKE, containing the opioid peptide endomorphin-2 (EM-2) and the NPFF receptor antagonist RF9. Our data obtained from the in vitro cyclic adenosine monophosphate experiment demonstrated that EKR functioned as a mixed mu-, delta-opioid receptors agonist and NPFF1 receptor antagonist/NPFF2 receptor partial agonist, whereas RKE acted as a multi-functional peptidomimetic with the mu-opioid agonism and the NPFF1 antagonism/NPFF2 partial agonism. Furthermore, EKR and RKE completely blocked the NPFF2 receptor-mediated neurite outgrowth of Neuro 2A cells. In vivo antinociception studies found that supraspinal administration of EKR and RKE dose-dependently produced potent antinociception via the mu-opioid receptor in the tail-flick test. In carrageenan inflammatory pain model, spinal administration of EKR and RKE induced dose-related analgesia, which was significantly reduced by the opioid antagonist naloxone and the NPFF antagonist RF9. Notably, compared with morphine, intracerebroventricular repeated administration of EKR and RKE maintained prolonged antinociceptive effectiveness. In addition, at the antinociceptive doses, these two branched peptidomimetics did not significantly inhibit gastrointestinal transit. Taken together, the present work suggest that EKR and RKE behave as multi-functional ligands with the opioid agonism and the NPFF1 antagonism/NPFF2 partial agonism, and produce prolonged antinociception with limited side effects. Moreover, our results imply that EKR and RKE might be interesting pharmacological tools for further investigating the biological function of the NPFF and opioid systems.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Discovery , Peptidomimetics/pharmacology , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/therapeutic use , Animals , Cell Line, Tumor , Drug Partial Agonism , Gastrointestinal Transit/drug effects , HEK293 Cells , Humans , Hyperalgesia/drug therapy , Male , Mice , Morphine/pharmacology , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Neuronal Outgrowth/drug effects , Peptidomimetics/chemical synthesis , Peptidomimetics/therapeutic use , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism
13.
Lab Invest ; 98(10): 1276-1290, 2018 10.
Article in English | MEDLINE | ID: mdl-29540859

ABSTRACT

Oxidative stress resulting from excessive production of reactive oxygen species (ROS) or impaired antioxidant defenses is closely related to the development of diabetic vascular complications, including nephropathy and atherosclerosis. Chronic activation of Janus kinase/Signal transducer and activator of transcription (JAK/STAT) signaling pathway contributes to diabetic complications by inducing expression of genes involved in cell proliferation, fibrosis, inflammation, and oxidative stress. Suppressors of cytokine signaling (SOCS) family of endogenous JAK/STAT regulators is an attractive target for therapeutic intervention. We investigated the beneficial effect of two different SOCS1-targeted therapies (adenovirus-mediated gene transfer and kinase-inhibitory region peptidomimetic) to combat oxidative stress injury in an experimental diabetes model of concomitant renal and macrovascular disease (streptozotocin-induced diabetic apolipoprotein E-deficient mouse). Diabetes resulted in progressive alteration of redox balance in mice, as demonstrated by increased ROS levels and decreased antioxidant activity, which ultimately led to renal dysfunction and vascular injury. The molecular and pathological alterations in early diabetes were partially reversed by preventive intervention with SOCS1-targeted therapies. Importantly, SOCS1 peptidomimetic provided reno- and atheroprotection in diabetic mice even in a setting of established disease. Compared with untreated controls, kidney and aorta from SOCS1-treated mice exhibited significantly lower levels of superoxide anion, DNA oxidation marker and NADPH oxidase (Nox) subunits, along with higher expression of antioxidant enzymes. These trends correlated with a reduction in parameters of renal damage (albuminuria, creatinine and tubular injury), atherosclerosis (lesion size) and inflammation (leukocytes and chemokines). Mechanistic studies in renal, vascular and phagocytic cells exposed to cytokines and high-glucose showed that SOCS1 blocked ROS generation by inhibiting both Nox complex assembly and Nox subunit expression, an effect mediated by inactivation of JAK2, STAT1, and PI3K signaling pathways. This study provides evidence for SOCS1-targeted therapies, especially SOCS1 peptidomimetic, as an alternative antioxidant strategy to limit the progression of diabetic micro- and macrovascular complications.


Subject(s)
Diabetic Angiopathies/therapy , Diabetic Nephropathies/therapy , Oxidative Stress , Suppressor of Cytokine Signaling 1 Protein/physiology , Animals , Antioxidants/metabolism , Aorta/metabolism , Genetic Therapy , Kidney/metabolism , Male , Mice , NADPH Oxidases/metabolism , Peptidomimetics/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Reactive Oxygen Species/metabolism , STAT1 Transcription Factor/metabolism
14.
J Pept Sci ; 24(6): e3084, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29870122

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune inflammatory disease. Early diagnosis can prevent joint erosion. However, available biomarkers do not always allow for clear distinction between RA and non-RA individuals. It has become known that bacteria/viruses are among the environmental triggers that initiate RA via multiple molecular mechanisms. Thus, to better understand the role of bacteria in RA, we synthetized 6 peptidomimetics of bacterial ureases' flap region. These peptides were then used to distinguish RA patients from healthy people sera by immunoblotting. Most patients' sera were bound to peptidomimetic characteristic for Enterobacter sp. and Klebsiella sp. flap urease. We also found similarities between peptidomimetic sequence and human proteins connected with RA. This pilot study suggests that bacteria may trigger RA via mechanism of molecular mimicry of urease to host proteins and ureases flap peptidomimetics may be potential candidate as a new additional diagnostic test.


Subject(s)
Arthritis, Rheumatoid/diagnosis , Peptidomimetics/therapeutic use , Urease/therapeutic use , Arthritis, Rheumatoid/pathology , Biomarkers/chemistry , Enterobacter/enzymology , Humans , Klebsiella/enzymology , Molecular Mimicry , Peptidomimetics/chemistry , Pilot Projects , Urease/chemistry
15.
Biochem J ; 474(1): 47-50, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28008090

ABSTRACT

Treatments for diabetes and obesity based on enteroendocrine hormones are a focus of research interest, partly due to the successes of glucagon-like peptide-1 (GLP-1) mimetic peptides in the treatment of diabetes and the correlation of altered enteroendocrine profiles with the positive metabolic outcomes of gastric bypass surgery. It is thought that simultaneous stimulation of more than one receptor might mimic the superior efficacy of the latter and dual or triple-agonist peptides are under investigation. An important step in developing multiple agonists is to establish the relative pharmacological potency and efficacy of new molecules at its different target receptors, and to optimise the balance of activities to achieve the desired treatment outcome. In a recent issue of the Biochemical Journal, Naylor et al. described how they used CRISPR technology to modulate endogenous receptor density in insulinoma cells to get the balance right for a dual incretin peptide engaging both GLP-1- and glucose-dependent insulinotropic polypeptide-receptors.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Diabetes Mellitus , Glucagon-Like Peptide 1 , Obesity , Animals , Cell Line, Tumor , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/therapy , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/therapeutic use , Humans , Obesity/genetics , Obesity/metabolism , Obesity/therapy , Peptidomimetics/therapeutic use
16.
J Am Soc Nephrol ; 28(2): 575-585, 2017 02.
Article in English | MEDLINE | ID: mdl-27609616

ABSTRACT

Diabetes is the main cause of CKD and ESRD worldwide. Chronic activation of Janus kinase and signal transducer and activator of transcription (STAT) signaling contributes to diabetic nephropathy by inducing genes involved in leukocyte infiltration, cell proliferation, and extracellular matrix accumulation. This study examined whether a cell-permeable peptide mimicking the kinase-inhibitory region of suppressor of cytokine signaling-1 (SOCS1) regulatory protein protects against nephropathy by suppressing STAT-mediated cell responses to diabetic conditions. In a mouse model combining hyperglycemia and hypercholesterolemia (streptozotocin diabetic, apoE-deficient mice), renal STAT activation status correlated with the severity of nephropathy. Notably, compared with administration of vehicle or mutant inactive peptide, administration of the SOCS1 peptidomimetic at either early or advanced stages of diabetes ameliorated STAT activity and resulted in reduced serum creatinine level, albuminuria, and renal histologic changes (mesangial expansion, tubular injury, and fibrosis) over time. Mice treated with the SOCS1 peptidomimetic also exhibited reduced kidney leukocyte recruitment (T lymphocytes and classic M1 proinflammatory macrophages) and decreased expression levels of proinflammatory and profibrotic markers that were independent of glycemic and lipid changes. In vitro, internalized peptide suppressed STAT activation and target gene expression induced by inflammatory and hyperglycemic conditions, reduced migration and proliferation in mesangial and tubuloepithelial cells, and altered the expression of cytokine-induced macrophage polarization markers. In conclusion, our study identifies SOCS1 mimicking as a feasible therapeutic strategy to halt the onset and progression of renal inflammation and fibrosis in diabetic kidney disease.


Subject(s)
Diabetic Nephropathies/drug therapy , Peptidomimetics/therapeutic use , Suppressor of Cytokine Signaling 1 Protein/therapeutic use , Animals , Disease Progression , Male , Mice , Suppressor of Cytokine Signaling 1 Protein/physiology
17.
Int J Mol Sci ; 19(9)2018 Sep 11.
Article in English | MEDLINE | ID: mdl-30208640

ABSTRACT

Inflammation is a physiological mechanism used by organisms to defend themselves against infection, restoring homeostasis in damaged tissues. It represents the starting point of several chronic diseases such as asthma, skin disorders, cancer, cardiovascular syndrome, arthritis, and neurological diseases. An increasing number of studies highlight the over-expression of inflammatory molecules such as oxidants, cytokines, chemokines, matrix metalloproteinases, and transcription factors into damaged tissues. The treatment of inflammatory disorders is usually linked to the use of unspecific small molecule drugs that can cause undesired side effects. Recently, many efforts are directed to develop alternative and more selective anti-inflammatory therapies, several of them imply the use of peptides. Indeed, peptides demonstrated as elected lead compounds toward several targets for their high specificity as well as recent and innovative synthetic strategies. Several endogenous peptides identified during inflammatory responses showed anti-inflammatory activities by inhibiting, reducing, and/or modulating the expression and activity of mediators. This review aims to discuss the potentialities and therapeutic use of peptides as anti-inflammatory agents in the treatment of different inflammation-related diseases and to explore the importance of peptide-based therapies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Inflammation/drug therapy , Peptides/therapeutic use , Peptidomimetics/therapeutic use , Amino Acid Sequence , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Autoimmune Diseases/complications , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Humans , Inflammation/complications , Inflammation/immunology , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/immunology , Nervous System Diseases/complications , Nervous System Diseases/drug therapy , Nervous System Diseases/immunology , Peptides/chemistry , Peptides/pharmacology , Peptidomimetics/chemistry , Peptidomimetics/pharmacology
19.
Biochim Biophys Acta Biomembr ; 1859(4): 577-585, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27580024

ABSTRACT

Membrane proteins play the central roles in a variety of cellular processes, ranging from nutrient uptake and signalling, to cell-cell communication. Their biological functions are directly related to how they fold and assemble; defects often lead to disease. Protein-protein interactions (PPIs) within the membrane are therefore of great interest as therapeutic targets. Here we review the progress in the application of membrane-insertable peptides for the disruption or stabilization of membrane-based PPIs. We describe the design and preparation of transmembrane peptide mimics; and of several categories of peptidomimetics used for study, including d-enantiomers, non-natural amino acids, peptoids, and ß-peptides. Further aspects of the review describe modifications to membrane-insertable peptides, including lipidation and cyclization via hydrocarbon stapling. These approaches provide a pathway toward the development of metabolically stable, non-toxic, and efficacious peptide modulators of membrane-based PPIs. This article is part of a Special Issue entitled: Lipid order/lipid defects and lipid-control of protein activity edited by Dirk Schneider.


Subject(s)
Membrane Lipids/chemistry , Membrane Proteins/chemistry , Peptides, Cyclic/chemistry , Peptidomimetics/chemistry , Peptoids/chemistry , Amino Acid Motifs , Asthma/drug therapy , Asthma/metabolism , Asthma/pathology , Cell Membrane/chemistry , Cell Membrane/metabolism , Drug Design , Humans , Membrane Lipids/antagonists & inhibitors , Membrane Lipids/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Peptides, Cyclic/metabolism , Peptides, Cyclic/therapeutic use , Peptidomimetics/metabolism , Peptidomimetics/therapeutic use , Peptoids/metabolism , Peptoids/therapeutic use , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Folding , Stereoisomerism
20.
Int J Med Sci ; 14(13): 1342-1359, 2017.
Article in English | MEDLINE | ID: mdl-29200948

ABSTRACT

Dengue is an important global threat caused by dengue virus (DENV) that records an estimated 390 million infections annually. Despite the availability of CYD-TDV as a commercial vaccine, its long-term efficacy against all four dengue virus serotypes remains unsatisfactory. There is therefore an urgent need for the development of antiviral drugs for the treatment of dengue. Peptide was once a neglected choice of medical treatment but it has lately regained interest from the pharmaceutical industry following pioneering advancements in technology. In this review, the design of peptide drugs, antiviral activities and mechanisms of peptides and peptidomimetics (modified peptides) action against dengue virus are discussed. The development of peptides as inhibitors for viral entry, replication and translation is also described, with a focus on the three main targets, namely, the host cell receptors, viral structural proteins and viral non-structural proteins. The antiviral peptides designed based on these approaches may lead to the discovery of novel anti-DENV therapeutics that can treat dengue patients.


Subject(s)
Dengue Virus/drug effects , Dengue/drug therapy , Peptides/therapeutic use , Peptidomimetics/therapeutic use , Antiviral Agents/therapeutic use , Dengue/epidemiology , Dengue/virology , Dengue Vaccines/therapeutic use , Dengue Virus/pathogenicity , Humans , Virus Internalization/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL