Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int Rev Neurobiol ; 170: 267-305, 2023.
Article in English | MEDLINE | ID: mdl-37741694

ABSTRACT

Chemotherapy has a significant positive impact in cancer treatment outcomes, reducing recurrence and mortality. However, many cancer surviving children and adults suffer from aberrant chemotherapy neurotoxic effects on learning, memory, attention, executive functioning, and processing speed. This chemotherapy-induced cognitive impairment (CICI) is referred to as "chemobrain" or "chemofog". While the underlying mechanisms mediating CICI are still unclear, there is strong evidence that chemotherapy accelerates the biological aging process, manifesting as effects which include telomere shortening, epigenetic dysregulation, oxidative stress, mitochondrial defects, impaired neurogenesis, and neuroinflammation, all of which are known to contribute to increased anxiety and neurocognitive decline. Despite the increased prevalence of CICI, there exists a lack of mechanistic understanding by which chemotherapy detrimentally affects cognition in cancer survivors. Moreover, there are no approved therapeutic interventions for this condition. To address this gap in knowledge, this review attempts to identify how adenosine signaling, particularly through the adenosine A2A receptor, can be an essential tool to attenuate accelerated aging phenotypes. Importantly, the adenosine A2A receptor uniquely stands at the crossroads of cancer treatment and improved cognition, given that it is widely known to control tumor induced immunosuppression in the tumor microenvironment, while also posited to be an essential regulator of cognition in neurodegenerative disease. Consequently, we propose that the adenosine A2A receptor may provide a multifaceted therapeutic strategy to enhance anticancer activity, while combating chemotherapy induced cognitive deficits, both which are essential to provide novel therapeutic interventions against accelerated aging in cancer survivors.


Subject(s)
Aging, Premature , Antineoplastic Agents , Cancer Survivors , Chemotherapy-Related Cognitive Impairment , Neoplasms , Neurodegenerative Diseases , Adult , Child , Humans , Adenosine , Chemotherapy-Related Cognitive Impairment/prevention & control , Neoplasms/drug therapy , Receptor, Adenosine A2A , Aging, Premature/chemically induced , Antineoplastic Agents/adverse effects
2.
Proc Natl Acad Sci U S A ; 119(28): e2206415119, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35867768

ABSTRACT

Chemotherapy-induced cognitive impairment (CICI) has emerged as a significant medical problem without therapeutic options. Using the platinum-based chemotherapy cisplatin to model CICI, we revealed robust elevations in the adenosine A2A receptor (A2AR) and its downstream effectors, cAMP and CREB, by cisplatin in the adult mouse hippocampus, a critical brain structure for learning and memory. Notably, A2AR inhibition by the Food and Drug Administration-approved A2AR antagonist KW-6002 prevented cisplatin-induced impairments in neural progenitor proliferation and dendrite morphogenesis of adult-born neurons, while improving memory and anxiety-like behavior, without affecting tumor growth or cisplatin's antitumor activity. Collectively, our study identifies A2AR signaling as a key pathway that can be therapeutically targeted to prevent cisplatin-induced cognitive impairments.


Subject(s)
Adenosine A2 Receptor Antagonists , Antineoplastic Agents , Chemotherapy-Related Cognitive Impairment , Cisplatin , Neurogenesis , Purines , Receptor, Adenosine A2A , Adenosine A2 Receptor Antagonists/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Chemotherapy-Related Cognitive Impairment/prevention & control , Cisplatin/adverse effects , Cognition/drug effects , Hippocampus/drug effects , Hippocampus/physiopathology , Mice , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Neural Stem Cells/physiology , Neurogenesis/drug effects , Purines/administration & dosage , Purines/therapeutic use , Receptor, Adenosine A2A/metabolism
3.
Eur Rev Med Pharmacol Sci ; 25(18): 5807-5817, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34604972

ABSTRACT

The term "chemobrain" refers to the cognitive dysfunction that occurs after chemotherapy, and it is also known as chemotherapy-induced cognitive impairment or "chemofog". The aim of this review is to bring together the findings of existing literature on the topic and summarize the current knowledge on the potential mechanisms of chemobrain. According to the reviewed studies, the mechanisms by which chemotherapy could cause chemobrain include disruption of hippocampal cell proliferation and neurogenesis, hormonal changes, increased oxidative stress and reactive oxygen species production, chronic increase in inflammation, and alterations in synaptic plasticity and long-term potentiation. While the effects of inflammation and oxidative stress on neurogenesis and their role in chemotherapy-induced cognitive impairment have been widely studied, the chemotherapy-induced cognitive impairment mechanisms that involve mitochondrial dysfunction, estrogen dysregulation, and increased transglutaminase 2 are still unclear. Further studies on these mechanisms are necessary to understand the effects of chemotherapy at the cellular and molecular level and facilitate the development of preventive and therapeutic strategies against chemotherapy-associated cognitive impairment or chemobrain.


Subject(s)
Chemotherapy-Related Cognitive Impairment/etiology , Cell Proliferation , Chemotherapy-Related Cognitive Impairment/prevention & control , Estrogens/metabolism , Hippocampus/cytology , Hippocampus/pathology , Humans , Inflammation , Mitochondria , Neurogenesis , Oxidative Stress , Protein Glutamine gamma Glutamyltransferase 2/metabolism , Reactive Oxygen Species
4.
Neurotherapeutics ; 18(3): 2107-2125, 2021 07.
Article in English | MEDLINE | ID: mdl-34312765

ABSTRACT

Although doxorubicin (Dox) is an effective chemotherapy medication used extensively in the treatment of breast cancer, it frequently causes debilitating neurological deficits known as chemobrain. Donepezil (DPZ), an acetylcholinesterase inhibitor, provides therapeutic benefits in various neuropathological conditions. However, comprehensive mechanistic insights regarding the neuroprotection of DPZ on cognition and brain pathologies in a Dox-induced chemobrain model remain obscure. Here, we demonstrated that Dox-treated rats manifested conspicuous cognitive deficits and developed chemobrain pathologies as indicated by brain inflammatory and oxidative insults, glial activation, defective mitochondrial homeostasis, increased potential lesions associated with Alzheimer's disease, disrupted neurogenesis, loss of dendritic spines, and ultimately neuronal death through both apoptosis and necroptosis. Intervention with DPZ co-treatment completely restored cognitive function by attenuating these pathological conditions induced by DOX. We also confirmed that DPZ treatment does not affect the anti-cancer efficacy of Dox in breast cancer cells. Together, our findings suggest that DPZ treatment confers potential neuroprotection against Dox-induced chemobrain.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Chemotherapy-Related Cognitive Impairment/prevention & control , Donepezil/therapeutic use , Doxorubicin/toxicity , Inflammation Mediators/antagonists & inhibitors , Oxidative Stress/drug effects , Animals , Chemotherapy-Related Cognitive Impairment/metabolism , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/therapeutic use , Donepezil/pharmacology , Female , Humans , Inflammation Mediators/metabolism , MCF-7 Cells , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress/physiology , Rats , Rats, Wistar , Treatment Outcome
6.
Article in English | MEDLINE | ID: mdl-32727341

ABSTRACT

Pharmacological therapy targeting the HER2 protein is one of the major breakthroughs in the treatment of cancer patients overexpressing HER2 who have increased survival rates. Despite improved survival, it is important to determine the less frequent adverse effects in order to tailor treatments more personalized to the patients' features. The possible impact of cancer treatments on cognitive functions is huge, and the effects of anti-HER 2 therapies on this issue have not been reviewed and are the objective of this study. Analysis of PubMed, Scopus, Cochrane library and Web of Science databases revealed six studies performed in breast and serous uterine cancer patients analyzing cognitive function under chemotherapy regimens including anti-HER2 drugs. Four of these studies reported small to significant worsening of cognitive function following chemotherapy regimens containing trastuzumab (the most widely used anti-HER2 drug). In neoadjuvant settings, and in breast cancer patients, treatment with the new anti-HER-2 drug trastuzumab emtansine seems to induce less cognitive impairment than therapeutic regimens containing chemotherapy and trastuzumab. Acute administration of trastuzumab induced cognitive impairment in gastric cancer mice models, confirming its ability to alter cognitive function in patients. More studies analyzing the impact of anti-HER2 therapy on cognitive function are necessary at preclinical and clinical levels in order to personalize pharmacological treatment and offer cancer patients a better quality of life.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Breast Neoplasms/drug therapy , Chemotherapy-Related Cognitive Impairment/etiology , Cognition/drug effects , Receptor, ErbB-2/antagonists & inhibitors , Trastuzumab/adverse effects , Animals , Breast Neoplasms/metabolism , Chemotherapy-Related Cognitive Impairment/diagnosis , Chemotherapy-Related Cognitive Impairment/prevention & control , Chemotherapy-Related Cognitive Impairment/psychology , Female , Humans , Quality of Life , Receptor, ErbB-2/metabolism , Risk Assessment , Risk Factors , Treatment Outcome
7.
Neuropharmacology ; 181: 108334, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33011199

ABSTRACT

Chemotherapy-induced cognitive dysfunction (chemobrain) is one of the major complaints for cancer patients treated with chemotherapy such as Doxorubicin (DOX). The induction of oxidative stress and neuroinflammation were identified as major contributors to such adverse effect. Caffeic acid phenethyl ester (CAPE) is a natural polyphenolic compound, that exhibits unique context-dependent antioxidant activity. It exhibits pro-oxidant effects in cancer cells, while it is a potent antioxidant and cytoprotective in normal cells. The present study was designed to investigate the potential neuroprotective effects of CAPE against DOX-induced cognitive impairment. Chemobrain was induced in Sprague Dawley rats via systemic DOX administration once per week for 4 weeks (2 mg/kg/week, i.p.). CAPE was administered at 10 or 20 µmol/kg/day, i.p., 5 days per week for 4 weeks. Morris water maze (MWM) and passive avoidance tests were used to assess learning and memory functions. Oxidative stress was evaluated via the colorimetric determination of GSH and MDA levels in both hippocampal and prefrontal cortex brain regions. However, inflammatory markers, acetylcholine levels, and neuronal cell apoptosis were assessed in the same brain areas using immunoassays including either ELISA, western blotting or immunohistochemistry. DOX produced significant impairment in learning and memory as indicated by the data generated from MWM and step-through passive avoidance tests. Additionally DOX-triggered oxidative stress as evidenced from the reduction in GSH levels and increased lipid peroxidation. Treatment with DOX resulted in neuroinflammation as indicated by the increase in NF-kB (p65) nuclear translocation in addition to boosting the levels of pro-inflammatory mediators (COX-II/TNF-α) along with the increased levels of glial fibrillary acid protein (GFAP) in the tested tissues. Moreover, DOX reduced acetylcholine levels and augmented neuronal cell apoptosis as supported by the increased active caspase-3 levels. Co-treatment with CAPE significantly counteracted DOX-induced behavioral and molecular abnormalities in rat brain tissues. Our results provide the first preclinical evidence for CAPE promising neuroprotective activity against DOX-induced neurodegeneration and memory deficits.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Caffeic Acids/therapeutic use , Chemotherapy-Related Cognitive Impairment/prevention & control , Doxorubicin/antagonists & inhibitors , Encephalitis/prevention & control , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Phenylethyl Alcohol/analogs & derivatives , Animals , Avoidance Learning/drug effects , Brain Chemistry , Caspase 3/metabolism , Chemotherapy-Related Cognitive Impairment/psychology , Doxorubicin/toxicity , Encephalitis/chemically induced , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation Mediators/metabolism , Male , Maze Learning/drug effects , Memory/drug effects , Motor Activity/drug effects , Phenylethyl Alcohol/therapeutic use , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley
8.
Curr Neuropharmacol ; 18(9): 838-851, 2020.
Article in English | MEDLINE | ID: mdl-32091339

ABSTRACT

BACKGROUND: Cognitive impairment is an adverse reaction of cancer chemotherapy and is likely to affect up to 75% of patients during the treatment and 35% of patients experience it for several months after the chemotherapy. Patients manifest symptoms like alteration in working ability, awareness, concentration, visual-verbal memory, attention, executive functions, processing speed, fatigue and behavioural dysfunctions. Post-chemotherapy, cancer survivors have a reduced quality of life due to the symptoms of chemobrain. Apart from this, there are clinical reports which also associate mood disorders, vascular complications, and seizures in some cases. Therefore, the quality of lifestyle of cancer patients/ survivors is severely affected and only worsens due to the absence of any efficacious treatments. With the increase in survivorship, it's vital to identify effective strategies, until then only symptomatic relief for chemobrain can be provided. The depressive symptoms were causally linked to the pathophysiological imbalance between the pro and antiinflammatory cytokines. CONCLUSION: The common causative factor, cytokines can be targeted for the amelioration of an associated symptom of both depression and chemotherapy. Thus, antidepressants can have a beneficial effect on chemotherapy-induced inflammation and cognitive dysfunction via cytokine balance. Also, neurogenesis property of certain antidepressant drugs rationalises their evaluation against CICI. This review briefly glances upon chemotherapy-induced cognitive impairment (CICI), and the modulatory effect of antidepressants on CICI pathomechanisms.


Subject(s)
Antidepressive Agents/therapeutic use , Antineoplastic Agents/adverse effects , Chemotherapy-Related Cognitive Impairment/physiopathology , Cognitive Dysfunction/chemically induced , Brain/drug effects , Chemotherapy-Related Cognitive Impairment/prevention & control , Cytokines , Drug Therapy , Drug-Related Side Effects and Adverse Reactions , Humans , Inflammation/chemically induced , Neurogenesis/drug effects , Oxidative Stress/drug effects , Quality of Life
9.
Biosci Biotechnol Biochem ; 84(6): 1201-1210, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31992173

ABSTRACT

Most breast cancer survivors receiving chemotherapy have severe cognitive impairment, often referred to as "chemobrain." Polydatin (PLD) is known to have many biological activities. Thus, this study aimed to determine whether symptoms of chemobrain can be prevented or relieved by PLD. The chemobrain models were established by intraperitoneal injection of doxorubicin (DOX, 2 mg/kg) in rats once a week for 4 weeks (DOX group and DOX+PLD group). In the PLD group and DOX+PLD group, PLD (50 mg/kg) was administered orally to rats every day. We found that PLD treatment significantly protected against DOX-induced learning and memory impairment, restored hippocampal histopathological architecture. Furthermore, PLD suppressed DOX-induced oxidative stress through up-regulating Nrf2, inhibited inflammatory response by activating the NF-κB pathway, and reduced hippocampal apoptosis. Therefore, the present study indicated that PLD offered neuroprotection against DOX-induced chemobrain. PLD may assist in preventing chemobrain after chemotherapy in patients with cancers.


Subject(s)
Antioxidants/administration & dosage , Apoptosis/drug effects , Chemotherapy-Related Cognitive Impairment/therapy , Drugs, Chinese Herbal/administration & dosage , Glucosides/administration & dosage , Oxidative Stress/drug effects , Phytotherapy/methods , Stilbenes/administration & dosage , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/adverse effects , Avoidance Learning/drug effects , Chemotherapy-Related Cognitive Impairment/etiology , Chemotherapy-Related Cognitive Impairment/prevention & control , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Fallopia japonica/chemistry , Hippocampus/drug effects , Hippocampus/pathology , Male , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spatial Memory/drug effects , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...