Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.759
Filter
1.
Adv Protein Chem Struct Biol ; 140: 199-248, 2024.
Article in English | MEDLINE | ID: mdl-38762270

ABSTRACT

The human gut microbiota is a complex and dynamic community of microorganisms, that influence metabolic, neurodevelopmental, and immune pathways. Microbial dysbiosis, characterized by changes in microbial diversity and relative abundances, is implicated in the development of various chronic neurological and neurodegenerative disorders. These disorders are marked by the accumulation of pathological protein aggregates, leading to the progressive loss of neurons and behavioural functions. Dysregulations in protein-protein interaction networks and signalling complexes, critical for normal brain function, are common in neurological disorders but challenging to unravel, particularly at the neuron and synapse-specific levels. To advance therapeutic strategies, a deeper understanding of neuropathogenesis, especially during the progressive disease phase, is needed. Biomarkers play a crucial role in identifying disease pathophysiology and monitoring disease progression. Proteomics, a powerful technology, shows promise in accelerating biomarker discovery and aiding in the development of novel treatments. In this chapter, we provide an in-depth overview of how proteomic techniques, utilizing various biofluid samples from patients with neurological conditions and diverse animal models, have contributed valuable insights into the pathogenesis of numerous neurological disorders. We also discuss the current state of research, potential challenges, and future directions in proteomic approaches to unravel neuro-pathological conditions.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Proteomics , Humans , Dysbiosis/metabolism , Dysbiosis/microbiology , Nervous System Diseases/metabolism , Nervous System Diseases/microbiology , Animals , Brain-Gut Axis , Biomarkers/metabolism
2.
Neurochem Int ; 177: 105771, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38761853

ABSTRACT

For most diseases and disorders occurring in the brain, the full causes behind them are yet unknown, but many show signs of dysfunction of amino acid transporters or abnormalities in amino acid metabolism. The blood-brain barrier (BBB) plays a key role in supporting the function of the central nervous system (CNS). Because of its unique structure, the BBB can maintain the optimal environment for CNS by controlling the passage of hydrophilic molecules from blood to the brain. Nutrients, such as amino acids, can cross the BBB via specific transporters. Many amino acids are essential for CNS function, and dysfunction of these amino acid transporters can lead to abnormalities in amino acid levels. This has been linked to causes behind certain genetic brain diseases, such as schizophrenia, autism spectrum disorder, and Huntington's disease (HD). One example of crucial amino acids is L-Cys, the rate-limiting factor in the biosynthesis of an important antioxidant, glutathione (GSH). Deficiency of L-Cys and GSH has been linked to oxidative stress and has been shown as a plausible cause behind certain CNS diseases, like schizophrenia and HD. This review presents the current status of potential L-Cys therapies and gives future directions that can be taken to improve amino acid transportation related to distinct CNS diseases.


Subject(s)
Amino Acid Transport Systems , Cysteine , Nervous System Diseases , Neuroprotective Agents , Humans , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Animals , Cysteine/metabolism , Nervous System Diseases/metabolism , Nervous System Diseases/drug therapy , Amino Acid Transport Systems/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects
3.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731911

ABSTRACT

In drug discovery, selecting targeted molecules is crucial as the target could directly affect drug efficacy and the treatment outcomes. As a member of the CCN family, CTGF (also known as CCN2) is an essential regulator in the progression of various diseases, including fibrosis, cancer, neurological disorders, and eye diseases. Understanding the regulatory mechanisms of CTGF in different diseases may contribute to the discovery of novel drug candidates. Summarizing the CTGF-targeting and -inhibitory drugs is also beneficial for the analysis of the efficacy, applications, and limitations of these drugs in different disease models. Therefore, we reviewed the CTGF structure, the regulatory mechanisms in various diseases, and drug development in order to provide more references for future drug discovery.


Subject(s)
Connective Tissue Growth Factor , Drug Discovery , Humans , Connective Tissue Growth Factor/metabolism , Drug Discovery/methods , Animals , Neoplasms/drug therapy , Neoplasms/metabolism , Eye Diseases/drug therapy , Eye Diseases/metabolism , Fibrosis , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Gene Expression Regulation/drug effects
4.
Int J Immunopathol Pharmacol ; 38: 3946320241250293, 2024.
Article in English | MEDLINE | ID: mdl-38712748

ABSTRACT

BACKGROUND: Cell metabolism functions without a stop in normal and pathological cells. Different metabolic changes occur in the disease. Cell metabolism influences biochemical and metabolic processes, signaling pathways, and gene regulation. Knowledge regarding disease metabolism is limited. OBJECTIVE: The review examines the cell metabolism of glucose, nucleotides, and lipids during homeostatic and pathological conditions of neurotoxicity, neuroimmunological disease, Parkinson's disease, thymoma in myasthenia gravis, and colorectal cancer. METHODS: Data collection includes electronic databases, the National Center for Biotechnology Information, and Google Scholar, with several inclusion criteria: cell metabolism, glucose metabolism, nucleotide metabolism, and lipid metabolism in health and disease patients suffering from neurotoxicity, neuroinflammation, Parkinson's disease, thymoma in myasthenia gravis. The initial number of collected and analyzed papers is 250. The final analysis included 150 studies out of 94 selected papers. After the selection process, 62.67% remains useful. RESULTS AND CONCLUSION: A literature search shows that signaling molecules are involved in metabolic changes in cells. Differences between cancer and neuroimmunological diseases are present in the result section. Our finding enables insight into novel therapeutic targets and the development of scientific approaches for cancer and neurological disease onset, outcome, progression, and treatment, highlighting the importance of metabolic dysregulation. Current understanding, emerging research technologies and potential therapeutic interventions in metabolic programming is disucussed and highlighted.


Subject(s)
Glucose , Lipid Metabolism , Neoplasms , Nervous System Diseases , Nucleotides , Humans , Neoplasms/metabolism , Neoplasms/drug therapy , Nervous System Diseases/metabolism , Nucleotides/metabolism , Glucose/metabolism , Animals , Signal Transduction
5.
Biomed Pharmacother ; 175: 116688, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692060

ABSTRACT

Metabolic syndrome (MetS) is characterized by insulin resistance, hyperglycemia, excessive fat accumulation and dyslipidemia, and is known to be accompanied by neuropathological symptoms such as memory loss, anxiety, and depression. As the number of MetS patients is rapidly increasing globally, studies on the mechanisms of metabolic imbalance-related neuropathology are emerging as an important issue. Ca2+/calmodulin-dependent kinase II (CaMKII) is the main Ca2+ sensor and contributes to diverse intracellular signaling in peripheral organs and the central nervous system (CNS). CaMKII exerts diverse functions in cells, related to mechanisms such as RNA splicing, reactive oxygen species (ROS) generation, cytoskeleton, and protein-protein interactions. In the CNS, CaMKII regulates vascular function, neuronal circuits, neurotransmission, synaptic plasticity, amyloid beta toxicity, lipid metabolism, and mitochondrial function. Here, we review recent evidence for the role of CaMKII in neuropathologic issues associated with metabolic disorders.


Subject(s)
Amyloid beta-Peptides , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Lipid Metabolism , Nervous System Diseases , Neuronal Plasticity , Humans , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Neuronal Plasticity/physiology , Animals , Lipid Metabolism/physiology , Amyloid beta-Peptides/metabolism , Nervous System Diseases/metabolism , Nervous System Diseases/physiopathology , Metabolic Syndrome/metabolism , Metabolic Syndrome/physiopathology
6.
Synapse ; 78(4): e22301, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38819491

ABSTRACT

Neurological disorders (NDs) are diseases of the central and peripheral nervous systems that affect more than one billion people worldwide. The risk of developing an ND increases with age due to the vulnerability of the different organs and systems to genetic, environmental, and social changes that consequently cause motor and cognitive deficits that disable the person from their daily activities and individual and social productivity. Intrinsic factors (genetic factors, age, gender) and extrinsic factors (addictions, infections, or lifestyle) favor the persistence of systemic inflammatory processes that contribute to the evolution of NDs. Neuroinflammation is recognized as a common etiopathogenic factor of ND. The study of new pharmacological options for the treatment of ND should focus on improving the characteristic symptoms and attacking specific molecular targets that allow the delay of damage processes such as neuroinflammation, oxidative stress, cellular metabolic dysfunction, and deregulation of transcriptional processes. In this review, we describe the possible role of sodium phenylbutyrate (NaPB) in the pathogenesis of Alzheimer's disease, hepatic encephalopathy, aging, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis; in addition, we describe the mechanism of action of NaPB and its beneficial effects that have been shown in various in vivo and in vitro studies to delay the evolution of any ND.


Subject(s)
Nervous System Diseases , Phenylbutyrates , Humans , Phenylbutyrates/therapeutic use , Phenylbutyrates/pharmacology , Animals , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism
7.
Neurosci Biobehav Rev ; 161: 105694, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38678735

ABSTRACT

There has been a growing awareness of the need for scientific research to focus on somatic and mental comorbidities in recent years due to the emerging evidence showing their substantial overlap at numerous levels. In this special issue, initiated by members of the EU-funded PRIME consortium ("Prevention and Remediation of Insulin Multimorbidity in Europe; www.prime-study.eu), the focus is on the comorbidities of metabolic disturbances, especially related to insulin signalling dysregulation and mental and neurological disorders. Thus, while obesity, type 2 diabetes, and metabolic syndrome are commonly known to be insulin-related disorders, the last decades have shown that neurodegenerative disorders, such as Alzheimer's disease, as well as neurodevelopment disorders, such as obsessive-compulsive disorder (OCD), autism spectrum disorders (ASDs) and attention deficit / hyperactivity disorder (ADHD) also fall into this category. The special issue draws together a series of basic and clinical review articles that describe the current knowledge and future perspectives regarding insulin comorbidities across a multidisciplinary group of experts.


Subject(s)
Insulin , Mental Disorders , Nervous System Diseases , Signal Transduction , Animals , Humans , Mental Disorders/metabolism , Insulin/metabolism , Nervous System Diseases/metabolism , Signal Transduction/physiology , Disease Models, Animal
8.
Biochem Pharmacol ; 224: 116218, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38643906

ABSTRACT

Non-coding RNAs (ncRNAs) are an assorted collection of transcripts that are not translated into proteins. Since their discovery, ncRNAs have gained prominence as crucial regulators of various biological functions across diverse cell types and tissues, and their abnormal functioning has been implicated in disease. Notably, extensive research has focused on the relationship between microRNAs (miRNAs) and human cancers, although other types of ncRNAs, such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), are also emerging as significant contributors to human disease. In this review, we provide a comprehensive summary of our current knowledge regarding the roles of miRNAs, lncRNAs, and circRNAs in cancer and other major human diseases, particularly cancer, cardiovascular, neurological, and infectious diseases. Moreover, we discuss the potential utilization of ncRNAs as disease biomarkers and as targets for therapeutic interventions.


Subject(s)
Neoplasms , RNA, Untranslated , Humans , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Neoplasms/genetics , Neoplasms/therapy , Neoplasms/metabolism , Neoplasms/drug therapy , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/therapy , MicroRNAs/genetics , MicroRNAs/metabolism , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Nervous System Diseases/therapy
9.
Cell Death Dis ; 15(4): 268, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627382

ABSTRACT

Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) represents the initial tumor suppressor gene identified to possess phosphatase activity, governing various cellular processes including cell cycle regulation, migration, metabolic pathways, autophagy, oxidative stress response, and cellular senescence. Current evidence suggests that PTEN is critical for stem cell maintenance, self-renewal, migration, lineage commitment, and differentiation. Based on the latest available evidence, we provide a comprehensive overview of the mechanisms by which PTEN regulates activities of different stem cell populations and influences neurological disorders, encompassing autism, stroke, spinal cord injury, traumatic brain injury, Alzheimer's disease and Parkinson's disease. This review aims to elucidate the therapeutic impacts and mechanisms of PTEN in relation to neurogenesis or the stem cell niche across a range of neurological disorders, offering a foundation for innovative therapeutic approaches aimed at tissue repair and regeneration in neurological disorders. This review unravels novel therapeutic strategies for tissue restoration and regeneration in neurological disorders based on the regulatory mechanisms of PTEN on neurogenesis and the stem cell niche.


Subject(s)
Nervous System Diseases , Parkinson Disease , Humans , Stem Cells/metabolism , Nervous System Diseases/therapy , Nervous System Diseases/metabolism , Cell Proliferation , Parkinson Disease/metabolism , Cell Differentiation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism
10.
Front Biosci (Landmark Ed) ; 29(4): 142, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38682185

ABSTRACT

Innate lymphocytes, including microglial cells, astrocytes, and oligodendrocytes, play a crucial role in initiating neuroinflammatory reactions inside the central nervous system (CNS). The prime focus of this paper is on the involvement and interplay of neurons and glial cells in neurological disorders such as Alzheimer's Disease (AD), Autism Spectrum Disorder (ASD), epilepsy, and multiple sclerosis (MS). In this review, we explore the specific contributions of microglia and astrocytes and analyzes multiple pathways implicated in neuroinflammation and disturbances in excitatory and inhibitory processes. Firstly, we elucidate the mechanisms through which toxic protein accumulation in AD results in synaptic dysfunction and deregulation of the immune system and examines the roles of microglia, astrocytes, and hereditary factors in the pathogenesis of the disease. Secondly, we focus on ASD and the involvement of glial cells in the development of the nervous system and the formation of connections between neurons and investigates the genetic connections associated with these processes. Lastly, we also address the participation of glial cells in epilepsy and MS, providing insights into their pivotal functions in both conditions. We also tried to give an overview of seven different pathways like toll-like receptor signalling pathway, MyD88-dependent and independent pathway, etc and its relevance in the context with these neurological disorders. In this review, we also explore the role of activated glial cells in AD, ASD, epilepsy, and MS which lead to neuroinflammation. Even we focus on excitatory and inhibitory imbalance in all four neurological disorders as imbalance affect the proper functioning of neuronal circuits. Finally, this review concludes that there is necessity for additional investigation on glial cells and their involvement in neurological illnesses.


Subject(s)
Nervous System Diseases , Neuroglia , Neurons , Animals , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Astrocytes/metabolism , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Cell Communication , Epilepsy/genetics , Epilepsy/metabolism , Epilepsy/physiopathology , Microglia/metabolism , Multiple Sclerosis/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/physiopathology , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Neuroglia/metabolism , Neuroinflammatory Diseases/metabolism , Neurons/metabolism , Signal Transduction
11.
Expert Rev Mol Med ; 26: e11, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38682637

ABSTRACT

Long non-coding RNAs (lncRNAs) are progressively being perceived as prominent molecular agents controlling multiple aspects of neuronal (patho)physiology. Amongst these is the HOX transcript antisense intergenic RNA, often abbreviated as HOTAIR. HOTAIR epigenetically regulates its target genes via its interaction with two different chromatin-modifying agents; histone methyltransferase polycomb-repressive complex 2 and histone demethylase lysine-specific demethylase 1. Parenthetically, HOTAIR elicits trans-acting sponging function against multiple micro-RNA species. Oncological research studies have confirmed the pathogenic functions of HOTAIR in multiple cancer types, such as gliomas and proposed it as a pro-oncological lncRNA. In fact, its expression has been suggested to be a predictor of the severity/grade of gliomas, and as a prognostic biomarker. Moreover, a propound influence of HOTAIR in other aspects of brain heath and disease states is just beginning to be unravelled. The objective of this review is to recapitulate all the relevant data pertaining to the regulatory roles of HOTAIR in neuronal (patho)physiology. To this end, we discuss the pathogenic mechanisms of HOTAIR in multiple neuronal diseases, such as neurodegeneration, traumatic brain injury and neuropsychiatric disorders. Finally, we also summarize the results from the studies incriminating HOTAIR in the pathogeneses of gliomas and other brain cancers. Implications of HOTAIR serving as a suitable therapeutic target in neuropathologies are also discussed.


Subject(s)
RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , Animals , Prognosis , Epigenesis, Genetic , Biomarkers , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Nervous System Diseases/therapy , Nervous System Diseases/pathology , Glioma/genetics , Glioma/pathology , Glioma/therapy , Glioma/metabolism
12.
Chem Rev ; 124(9): 5470-5504, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38607675

ABSTRACT

Lysophosphatidylserine (lyso-PS) has emerged as yet another important signaling lysophospholipid in mammals, and deregulation in its metabolism has been directly linked to an array of human autoimmune and neurological disorders. It has an indispensable role in several biological processes in humans, and therefore, cellular concentrations of lyso-PS are tightly regulated to ensure optimal signaling and functioning in physiological settings. Given its biological importance, the past two decades have seen an explosion in the available literature toward our understanding of diverse aspects of lyso-PS metabolism and signaling and its association with human diseases. In this Review, we aim to comprehensively summarize different aspects of lyso-PS, such as its structure, biodistribution, chemical synthesis, and SAR studies with some synthetic analogs. From a biochemical perspective, we provide an exhaustive coverage of the diverse biological activities modulated by lyso-PSs, such as its metabolism and the receptors that respond to them in humans. We also briefly discuss the human diseases associated with aberrant lyso-PS metabolism and signaling and posit some future directions that may advance our understanding of lyso-PS-mediated mammalian physiology.


Subject(s)
Lysophospholipids , Signal Transduction , Humans , Lysophospholipids/metabolism , Lysophospholipids/chemistry , Animals , Autoimmune Diseases/metabolism , Nervous System Diseases/metabolism
13.
Nat Rev Neurol ; 20(5): 269-287, 2024 05.
Article in English | MEDLINE | ID: mdl-38609644

ABSTRACT

Neurofilament proteins have been validated as specific body fluid biomarkers of neuro-axonal injury. The advent of highly sensitive analytical platforms that enable reliable quantification of neurofilaments in blood samples and simplify longitudinal follow-up has paved the way for the development of neurofilaments as a biomarker in clinical practice. Potential applications include assessment of disease activity, monitoring of treatment responses, and determining prognosis in many acute and chronic neurological disorders as well as their use as an outcome measure in trials of novel therapies. Progress has now moved the measurement of neurofilaments to the doorstep of routine clinical practice for the evaluation of individuals. In this Review, we first outline current knowledge on the structure and function of neurofilaments. We then discuss analytical and statistical approaches and challenges in determining neurofilament levels in different clinical contexts and assess the implications of neurofilament light chain (NfL) levels in normal ageing and the confounding factors that need to be considered when interpreting NfL measures. In addition, we summarize the current value and potential clinical applications of neurofilaments as a biomarker of neuro-axonal damage in a range of neurological disorders, including multiple sclerosis, Alzheimer disease, frontotemporal dementia, amyotrophic lateral sclerosis, stroke and cerebrovascular disease, traumatic brain injury, and Parkinson disease. We also consider the steps needed to complete the translation of neurofilaments from the laboratory to the management of neurological diseases in clinical practice.


Subject(s)
Biomarkers , Intermediate Filaments , Nervous System Diseases , Neurofilament Proteins , Humans , Biomarkers/metabolism , Biomarkers/blood , Nervous System Diseases/diagnosis , Nervous System Diseases/metabolism , Nervous System Diseases/blood , Neurofilament Proteins/blood , Intermediate Filaments/metabolism
14.
FEBS Lett ; 598(9): 959-977, 2024 May.
Article in English | MEDLINE | ID: mdl-38644468

ABSTRACT

Reversible S-acylation plays a pivotal role in various biological processes, modulating protein functions such as subcellular localization, protein stability/activity, and protein-protein interactions. These modifications are mediated by acyltransferases and deacylases, among which the most abundant modification is S-palmitoylation. Growing evidence has shown that this rivalrous pair of modifications, occurring in a reversible cycle, is essential for various biological functions. Aberrations in this process have been associated with various diseases, including cancer, neurological disorders, and immune diseases. This underscores the importance of studying enzymes involved in acylation and deacylation to gain further insights into disease pathogenesis and provide novel strategies for disease treatment. In this Review, we summarize our current understanding of the structure and physiological function of deacylases, highlighting their pivotal roles in pathology. Our aim is to provide insights for further clinical applications.


Subject(s)
Neoplasms , Humans , Animals , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/genetics , Acyltransferases/metabolism , Acyltransferases/chemistry , Nervous System Diseases/enzymology , Nervous System Diseases/metabolism , Acylation , Lipoylation , Protein Processing, Post-Translational , Immune System Diseases/enzymology , Immune System Diseases/metabolism
15.
Cells ; 13(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38667285

ABSTRACT

Neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), stroke, and aneurysms, are characterized by the abnormal accumulation and aggregation of disease-causing proteins in the brain and spinal cord. Recent research suggests that proteins linked to these conditions can be secreted and transferred among cells using exosomes. The transmission of abnormal protein buildup and the gradual degeneration in the brains of impacted individuals might be supported by these exosomes. Furthermore, it has been reported that neuroprotective functions can also be attributed to exosomes in neurodegenerative diseases. The potential neuroprotective functions may play a role in preventing the formation of aggregates and abnormal accumulation of proteins associated with the disease. The present review summarizes the roles of exosomes in neurodegenerative diseases as well as elucidating their therapeutic potential in AD, PD, ALS, HD, stroke, and aneurysms. By elucidating these two aspects of exosomes, valuable insights into potential therapeutic targets for treating neurodegenerative diseases may be provided.


Subject(s)
Exosomes , Exosomes/metabolism , Humans , Animals , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Vascular Diseases/metabolism , Vascular Diseases/pathology , Nervous System Diseases/metabolism , Nervous System Diseases/pathology
16.
J Integr Neurosci ; 23(4): 86, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38682220

ABSTRACT

Due to the growth of the elderly population, age-related neurological disorders are an increasing problem. Aging begins very gradually and later leads to several neurological issues such as lower neurotransmitter levels, oxidative stress, neuronal inflammation, and continual neuronal loss. These changes might contribute to brain disorders such as Alzheimer's disease (AD), dementia or mild cognitive impairment, and epilepsy and glioma, and can also aggravate these disorders if they were previously present. Momordica charantia (bitter gourd), a member of the Cucurbitaceae family, is a good source of carbohydrates, proteins, vitamins, and minerals. It is used for diabetes and known for its hypoglycemic and antioxidant effects. In this review, we discuss the pharmaceutical effects of M. charantia on age-related neurological disorders. We searched several databases, including PubMed and Google Scholar, using MeSH terms. We searched articles published up until 2022 regardless of publication language. M. charantia is rich in luteolin, which increases acetylcholine in neurons by binding to enzymes in acetylcholine metabolism pathways, including butyrylcholinesterase and acetylcholinesterase. This binding inhibits the hyperphosphorylation of tau protein by restraining its kinase enzyme. Furthermore, this substance can lower serum cholesterol and has multi-target activity in AD and memory loss. M. charantia can also improve memory by decreasing tau protein and it also has potent antioxidant activity and anti-inflammatory effects. This review highlights that M. charantia has effects on many age-related neurological disorders, and can be a cost-effective supplement with minimal side effects.


Subject(s)
Momordica charantia , Momordica charantia/chemistry , Humans , Animals , Aging/drug effects , Aging/physiology , Aging/metabolism , Plant Extracts/pharmacology , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism
17.
Biochem Soc Trans ; 52(2): 553-565, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38563502

ABSTRACT

Given the current paucity of effective treatments in many neurological disorders, delineating pathophysiological mechanisms among the major psychiatric and neurodegenerative diseases may fuel the development of novel, potent treatments that target shared pathways. Recent evidence suggests that various pathological processes, including bioenergetic failure in mitochondria, can perturb the function of fast-spiking, parvalbumin-positive neurons (PV+). These inhibitory neurons critically influence local circuit regulation, the generation of neuronal network oscillations and complex brain functioning. Here, we survey PV+ cell vulnerability in the major neuropsychiatric, and neurodegenerative diseases and review associated cellular and molecular pathophysiological alterations purported to underlie disease aetiology.


Subject(s)
Mitochondria , Neurodegenerative Diseases , Neurons , Parvalbumins , Humans , Parvalbumins/metabolism , Neurodegenerative Diseases/metabolism , Mitochondria/metabolism , Animals , Neurons/metabolism , Nervous System Diseases/metabolism , Brain/metabolism
18.
J Cell Physiol ; 239(5): e31223, 2024 May.
Article in English | MEDLINE | ID: mdl-38530191

ABSTRACT

As an essential trace element for organisms, zinc participates in various physiological processes, such as RNA transcription, DNA replication, cell proliferation, and cell differentiation. The destruction of zinc homeostasis is associated with various diseases. Zinc homeostasis is controlled by the cooperative action of zinc transporter proteins that are responsible for the influx and efflux of zinc. Zinc transporter proteins are mainly categorized into two families: Zrt/Irt-like protein (SLC39A/ZIP) family and zinc transporter (SLC30A/ZNT) family. ZIP transporters contain 14 members, namely ZIP1-14, which can be further divided into four subfamilies. Currently, ZIP transporters-regulated zinc homeostasis is one of the research hotspots. Cumulative evidence suggests that ZIP transporters-regulated zinc homeostasis may cause physiological dysfunction and contribute to the onset and progression of diverse diseases, such as cancers, neurological diseases, and cardiovascular diseases. In this review, we initially discuss the structure and distribution of ZIP transporters. Furthermore, we comprehensively review the latest research progress of ZIP transporters-regulated zinc homeostasis in diseases, providing a new perspective into new therapeutic targets for treating related diseases.


Subject(s)
Cardiovascular Diseases , Cation Transport Proteins , Neoplasms , Nervous System Diseases , Zinc , Animals , Humans , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/genetics , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , Homeostasis/physiology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Zinc/metabolism , Nervous System Diseases/genetics , Nervous System Diseases/metabolism
19.
Clin Neurol Neurosurg ; 240: 108250, 2024 05.
Article in English | MEDLINE | ID: mdl-38552364

ABSTRACT

Hypoxia is a prevalent characteristic of numerous neurological disorders including stroke, Alzheimer's disease, and Parkinson's disease. Extracellular vesicles (EVs) are minute particles released by cells that contain diverse biological materials, including proteins, lipids, and nucleic acids. They have been implicated in a range of physiological and pathological processes including intercellular communication, immune responses, and disease progression. EVs are believed to play a pivotal role in modulating the microenvironment of hypoxia-associated neurological diseases. These EVs are capable of transporting hypoxia-inducible factors such as proteins and microRNAs to neighboring or remote cells, thereby influencing their behavior. Furthermore, EVs can traverse the blood-brain barrier, shielding the brain from detrimental substances in the bloodstream. This enables them to deliver their payload directly to the brain cells, potentially intensifying the effects of hypoxia. Nonetheless, the capacity of EVs to breach the blood-brain barrier presents new opportunities for drug delivery. The objective of this study was to elucidate the role of EVs as mediators of information exchange during tissue hypoxia, a pathophysiological process in ischemic stroke and malignant gliomas. We also investigated their involvement in the progression and regression of major diseases of the central nervous system, which are pertinent to the development of therapeutic interventions for neurological disorders.


Subject(s)
Extracellular Vesicles , Humans , Extracellular Vesicles/metabolism , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Hypoxia/metabolism , Blood-Brain Barrier/metabolism , Animals
20.
Pharmacol Res ; 203: 107149, 2024 May.
Article in English | MEDLINE | ID: mdl-38518830

ABSTRACT

Neuronal health is closely linked to the homeostasis of intracellular organelles, and organelle dysfunction affects the pathological progression of neurological diseases. In contrast to isolated cellular compartments, a growing number of studies have found that organelles are largely interdependent structures capable of communicating through membrane contact sites (MCSs). MCSs have been identified as key pathways mediating inter-organelle communication crosstalk in neurons, and their alterations have been linked to neurological disease pathology. The endoplasmic reticulum (ER) is a membrane-bound organelle capable of forming an extensive network of pools and tubules with important physiological functions within neurons. There are multiple MCSs between the ER and other organelles and the plasma membrane (PM), which regulate a variety of cellular processes. In this review, we focus on ER-organelle MCSs and their role in a variety of neurological diseases. We compared the biological effects between different tethering proteins and the effects of their respective disease counterparts. We also discuss how altered ER-organelle contacts may affect disease pathogenesis. Therefore, understanding the molecular mechanisms of ER-organelle MCSs in neuronal homeostasis will lay the foundation for the development of new therapies targeting ER-organelle contacts.


Subject(s)
Endoplasmic Reticulum , Nervous System Diseases , Signal Transduction , Humans , Endoplasmic Reticulum/metabolism , Animals , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Neurons/metabolism , Organelles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...