Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Opin Neurobiol ; 75: 102554, 2022 08.
Article in English | MEDLINE | ID: mdl-35649324

ABSTRACT

Macroautophagy (hereafter referred to as autophagy) is an essential quality-control pathway in neurons, which face unique functional and morphological challenges in maintaining the integrity of organelles and the proteome. To overcome these challenges, neurons have developed compartment-specific pathways for autophagy. In this review, we discuss the organization of the autophagy pathway, from autophagosome biogenesis, trafficking, to clearance, in the neuron. We dissect the compartment-specific mechanisms and functions of autophagy in axons, dendrites, and the soma. Furthermore, we highlight examples of how steps along the autophagy pathway are impaired in the context of aging and neurodegenerative disease, which underscore the critical importance of autophagy in maintaining neuronal function and survival.


Subject(s)
Neurodegenerative Diseases , Aging , Autophagy/physiology , Axons/physiology , Humans , Neurodegenerative Diseases/metabolism , Neurons/physiology
2.
J Am Chem Soc ; 143(8): 3086-3093, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33600171

ABSTRACT

An interruption in Aß homeostasis leads to the deposit of neurotoxic amyloid plaques and is associated with Alzheimer's disease. A supramolecular strategy based on the assembly of peptidomimetic agents into functional vesicles has been conceived for the simultaneous inhibition of Aß42 fibrillation and expedited clearance of Aß42 aggregates. Tris-pyrrolamide peptidomimetic, ADH-353, contains one hydrophobic N-butyl and two hydrophilic N-propylamine side chains and readily forms vesicles under physiological conditions. These vesicles completely rescue both mouse neuroblastoma N2a and human neuroblastoma SH-SY5Y cells from the cytotoxicity that follows from Aß42 misfolding likely in mitochondria. Biophysical studies, including confocal imaging, demonstrate the biocompatibility and selectivity of the approach toward this aberrant protein assembly in cellular milieu.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/toxicity , Peptide Fragments/chemistry , Peptide Fragments/toxicity , Peptidomimetics/pharmacology , Protein Aggregates/drug effects , Cell Line, Tumor , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Protein Folding/drug effects
3.
Org Biomol Chem ; 16(5): 733-741, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29303519

ABSTRACT

A library of N-substituted oligopyrrolamides was designed to modulate the aggregation kinetics of islet amyloid polypeptide (IAPP). IAPP is a hormonal peptide, co-secreted with insulin in the pancreatic ß-cells. IAPP samples a variety of conformations, starting from a native random coil to membrane-associated α-helical intermediates and eventually terminates in the amyloid plaques rich in ß-sheet structures. A growing body of evidence suggests that membrane bound α-helical intermediates are the key cytotoxic species that impair the functionality and viability of ß-cells and contribute to the onset of type 2 diabetes mellitus (DM2). The N-substituted oligopyrrolamides were screened against the aggregation of IAPP using amyloid kinetic assays. A tripyrrole, ADH-101, was the most effective antagonist of IAPP fibrillation in a physiologically relevant lipid membrane system as well as under de novo conditions. ADH-101 induces/stabilizes a secondary structure in IAPP which potentially affects its downstream functions. ADH-101 efficiently affects IAPP-mediated liposome leakage and cell toxicity in insulin secreting cells. ADH-101 inhibits the elongation process potentially binding to the monomeric IAPP and attenuating its access to the preformed fibers. More importantly, oligopyrrolamides are better inhibitors of IAPP aggregation than analogous oligopyridylamides and have more desirable biological properties reflected by their partition coefficients. In essence, an oligopyrrolamide scaffold has been designed which modulates the membrane bound helical intermediates of IAPP and affects their downstream functions such as oligomerization, membrane poration, and cytotoxicity.


Subject(s)
Islet Amyloid Polypeptide/chemistry , Protein Aggregates/drug effects , Protein Structure, Secondary/drug effects , Pyrroles/chemistry , Pyrroles/pharmacology , Amides/chemistry , Amides/pharmacology , Amyloid/chemistry , Amyloid/metabolism , Animals , Cell Line , Cell Survival/drug effects , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islet Amyloid Polypeptide/metabolism , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/metabolism , Rats
4.
FASEB J ; 31(5): 2168-2184, 2017 05.
Article in English | MEDLINE | ID: mdl-28183803

ABSTRACT

Overexpression of mitochondria-bound hexokinase II (HKII) in cancer cells plays an important role in their metabolic reprogramming and protects them against apoptosis, thereby facilitating their growth and proliferation. Here, we show that covalently coupling a peptide corresponding to the mitochondrial membrane-binding N-terminal 15 aa of HKII (pHK) to a short, penetration-accelerating sequence (PAS) enhances the cellular uptake, mitochondrial localization, and cytotoxicity of the peptide in HeLa cells. Further analysis revealed that pHK-PAS depolarized mitochondrial membrane potential, inhibited mitochondrial respiration and glycolysis, and depleted intracellular ATP levels. The effects of pHK-PAS were correlated with dissociation of endogenous full-length HKII from mitochondria and release of cytochrome c Of significance, pHK-PAS treatment of noncancerous HEK293 cells resulted in substantially lower cytotoxicity. Thus, pHK-PAS effectively disrupted the mitochondria-HKII association in cancer cells, which led to mitochondrial dysfunction and, finally, apoptosis. Our results demonstrate the potential of the pHK-PAS cell-penetrating peptide as a novel therapeutic strategy in cancer.-Woldetsadik, A. D., Vogel, M. C., Rabeh, W. M., Magzoub, M. Hexokinase II-derived cell-penetrating peptide targets mitochondria and triggers apoptosis in cancer cells.


Subject(s)
Apoptosis/physiology , Hexokinase/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Cell Line, Tumor , Cell-Penetrating Peptides/metabolism , Cytochromes c/metabolism , Glycolysis/physiology , HEK293 Cells , Humans
5.
Arch Biochem Biophys ; 613: 31-42, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27818203

ABSTRACT

Prion diseases are associated with conversion of cellular prion protein (PrPC) into an abnormally folded and infectious scrapie isoform (PrPSc). We previously showed that peptides derived from the unprocessed N-termini of mouse and bovine prion proteins, mPrP1-28 and bPrP1-30, function as cell-penetrating peptides (CPPs), and destabilize model membrane systems, which could explain the infectivity and toxicity of prion diseases. However, subsequent studies revealed that treatment with mPrP1-28 or bPrP1-30 significantly reduce PrPSc levels in prion-infected cells. To explain these seemingly contradictory results, we correlated the aggregation, membrane perturbation and cytotoxicity of the peptides with their cellular uptake and intracellular localization. Although the peptides have a similar primary sequence, mPrP1-28 is amyloidogenic, whereas bPrP1-30 forms smaller oligomeric or non-fibrillar aggregates. Surprisingly, bPrP1-30 induces much higher cytotoxicity than mPrP1-28, indicating that amyloid formation and toxicity are independent. The toxicity is correlated with prolonged residence at the plasma membrane and membrane perturbation. Both ordered aggregation and toxicity of the peptides are inhibited by low pH. Under non-toxic conditions, the peptides are internalized by lipid-raft dependent macropinocytosis and localize to acidic lysosomal compartments. Our results shed light on the antiprion mechanism of the prion protein-derived CPPs and identify a potential site for PrPSc formation.


Subject(s)
Amyloid/chemistry , Cell-Penetrating Peptides/chemistry , Prion Proteins/chemistry , Protein Sorting Signals , Animals , Binding Sites , Cattle , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Survival , Endosomes/chemistry , Endosomes/metabolism , HeLa Cells , Humans , Hydrogen-Ion Concentration , Kinetics , Liposomes/chemistry , Lysosomes/chemistry , Mice , Microscopy, Electron, Transmission , Peptides/chemistry , Protein Domains , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...