Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Mol Biol Rep ; 51(1): 930, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39174728

RESUMEN

BACKGROUND: Among the three most used anticancer drugs, cyclophosphamide, Adriamycin, and 5-Fluorouracil (CAF), the most significant outcome is chemobrain, caused by increased oxidative stress, inflammatory insult, and mitochondrial dysfunction. OBJECTIVE: In this study, endogenous antioxidant coenzyme Q10 (CoQ10) was evaluated for its neuroprotective effects in CICI. MATERIALS AND METHODS: The chemobrain was induced in Swiss albino female mice by administering CAF (40 + 4 + 25 mg/kg) intraperitoneal (i.p.) in three cycles (single injection per week) followed by treatment with CoQ10 (40 mg/kg; p.o.) for up to 3 weeks followed by behavioral, biochemical, molecular and histopathological analysis. RESULTS: Treatment with CoQ10 significantly improved cognition by improving exploring time in novel objects recognition test followed by increasing the time spent in the target quadrant in MWM test as compared to CAF-treated animals. Moreover, CoQ10 demonstrated antioxidant properties by reducing the expression of LPO while increasing levels of GSH, SOD, and catalase as compared to CAF-treated animals. While the levels of AChEs were significantly reduced after CoQ10 treatment in CAF-treated animals. In terms of its mechanism, it effectively counteracted the pro-inflammatory substances (TNF-α and IL-1ß) triggered by CAF while also enhancing the levels of anti-inflammatory markers (IL-10 and Nrf2). Moreover, CoQ10 showed mitochondrial enhancers and it improved the level of Complex (I, II, and IV). Besides that, mitochondrial morphological analysis was done by TEM, and neuronal morphology along with quantification analysis was performed by H&E staining using Image J software to confirm the neuroprotective effect of CoQ10 over CAF-induced cognitive impairment. CONCLUSION: This study suggests CoQ10 can protect the mitochondria by imposing antioxidant, and anti-inflammatory properties, which could be a potential therapy for CICI.


Asunto(s)
Antioxidantes , Estrés Oxidativo , Ubiquinona , Animales , Ubiquinona/análogos & derivados , Ubiquinona/farmacología , Ubiquinona/uso terapéutico , Ratones , Femenino , Antioxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Deterioro Cognitivo Relacionado con la Quimioterapia/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Doxorrubicina/efectos adversos , Fluorouracilo/efectos adversos , Fluorouracilo/farmacología , Modelos Animales de Enfermedad , Antineoplásicos/farmacología , Antineoplásicos/efectos adversos , Ciclofosfamida/efectos adversos , Ciclofosfamida/farmacología
2.
BMC Womens Health ; 24(1): 406, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39020328

RESUMEN

OBJECTIVE: To study the effects of chemotherapy on cognitive function in breast cancer patients, and to investigate the relationship of MemTrax test of memory and related functions to the FACT-Cog functional self-assessment for the evaluation and management of chemobrain. METHODS: In this prospective cohort study, clinical information of pathologically confirmed female breast cancer patients who decided to receive chemotherapy were collected in a questionnaire which was developed for this study and provided as a supplementary file. The FACT-Cog self-assessment and MemTrax test were administered before and after the chemotherapy treatments. Patients with chemobrain were identified using published criteria based on FACT-Cog scores, and MemTrax scores from chemobrain patients were analyzed. RESULTS: Fifty-six patients participated in this study, of which 41 participants completed 4 or more cycles of chemotherapy and were included in the final analyses here. Using the reported high end of minimal clinical differences (10.6 points) of FACT-Cog before and after chemotherapy, 18 patients suffered from chemobrain in this study. In these 18 chemobrain patients, no cognitive impairments were detected by MemTrax, which paradoxically demonstrated an improvement in the normal cognitive range. CONCLUSION: The cognitive impairment induced by chemotherapy in breast cancer patients is detectable by the FACT-Cog in a Chinese cohort but is not detected by the MemTrax memory test. The fact that the more objective MemTrax could not detect the impairment could alleviate patients' concerns which in turn would be beneficial for patients' mental health.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/psicología , Persona de Mediana Edad , Estudios Prospectivos , Adulto , Pruebas Neuropsicológicas/estadística & datos numéricos , Antineoplásicos/efectos adversos , Antineoplásicos/uso terapéutico , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Anciano , Memoria/efectos de los fármacos , Encuestas y Cuestionarios , Estudios de Cohortes
3.
J Mol Histol ; 55(4): 599-625, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39042217

RESUMEN

Today, we critically need alternative therapeutic options for chemotherapy-induced cognitive impairment (CICI), often known as chemo brain. Mitochondrial dysfunction and oxidative stress are two of the primary processes that contribute to the development of chemobrain. Therefore, the purpose of this study was to investigate how CoQ10 and berberine shield neurons from chemotherapy-induced damage in in-vitro studies and memory loss in vivo studies. For the in-vitro investigation, we employed SH-SY5Y cell lines, and for the in-vivo study, we used female Swiss albino mice divided into seven different groups. Data from in-vitro studies revealed that treatment with coenzyme Q10 (CoQ10) and berberine improved chemotherapy-induced toxicity by reducing mitochondrial and total cellular ROS, as well as apoptosis-elicited markers (caspase 3 and 9). CoQ10 and berberine therapy inhibited the nuclear translocation of NF-κB and, consequently, the subsequent expressions of NLRP3 and IL-1ß, implying the prevention of inflammasome formation. Furthermore, CoQ10 and berberine therapy boosted Nrf2 levels. This is a regulator for cellular resistance to oxidants. The in vivo results showed that treatment with CoQ10 (40 mg/kg) and berberine (200 mg/kg) improved the behavioral alterations induced by CAF (40/4/25 mg/kg) in both the Morris Water Maze (MWM) and Novel Object Recognition (NOR) tests. Furthermore, biochemical and molecular evidence revealed the antioxidant, mitochondrial restorative, and anti-inflammatory potential of CoQ10 (40 mg/kg) and berberine (200 mg/kg) against CAF (40/4/25 mg/kg) subjected mice. In addition, the histological analysis using H&E staining and transmission electron microscopy (for mitochondrial morphology) showed that mice treated with the cocktails had an increased number of healthy neurons with intact mitochondria and a reduced presence of autophagic vacuoles in the hippocampal region of the brain. These findings back up our theory about this novel cocktail method for CAF-induced cognitive impairment.


Asunto(s)
Berberina , Factor 2 Relacionado con NF-E2 , FN-kappa B , Transducción de Señal , Ubiquinona , Animales , FN-kappa B/metabolismo , Ratones , Transducción de Señal/efectos de los fármacos , Femenino , Factor 2 Relacionado con NF-E2/metabolismo , Berberina/farmacología , Ubiquinona/análogos & derivados , Ubiquinona/farmacología , Ubiquinona/administración & dosificación , Humanos , Línea Celular Tumoral , Estrés Oxidativo/efectos de los fármacos , Deterioro Cognitivo Relacionado con la Quimioterapia/metabolismo , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Apoptosis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo
4.
Neuropharmacology ; 254: 109981, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38704022

RESUMEN

The chemotherapeutic agent Doxorubicin (DOX) is known to cause chemotherapy-induced cognitive impairment (CICI). Maraviroc, a potent C-C chemokine receptor 5 (CCR5) antagonist, shows neuroprotective properties, while its role in CICI remains unclear. This study determined the therapeutic potential of maraviroc on CICI. Adult C57BL/6J mice with implanted breast cancer cells received four weekly intraperitoneal injections of saline (Control group), 5 mg/kg DOX (DOX group), 10 mg/kg maraviroc (MVC group), or 5 mg/kg DOX with 10 mg/kg maraviroc (DOX + MVC group). The Morris Water Maze (MWM) was used for neurobehavioural test. Western blot analysis and immunofluorescence were used to evaluate the expressions of inflammatory markers, apoptosis-related proteins, and synaptic-related proteins. The volume and weight of tumor were also evaluated after treatments. DOX treatment significantly increased chemokines (CCL3, CCL4) and inflammatory cytokines (IL-1ß, TNF-α) in tumor-bearing mice hippocampus. While maraviroc administration reduced hippocampal proinflammatory factors compared to the DOX group. Furthermore, it also lowered apoptosis markers, restored synaptic proteins levels, and inhibited the NF-κB/NLRP3 pathway. Accordingly, maraviroc treatment significantly improved DOX-induced neurobehavioural impairments as evidenced by an increased number of platform crossings and percentage of target quadrant time in the MWM test. Additionally, when combined with DOX, maraviroc had additional inhibitory effects on tumor growth. These findings suggest that maraviroc can mitigate DOX-induced CICI by suppressing elevated proinflammatory chemokines and cytokines through the NF-κB/NLRP3 pathway, potentially offering an anti-tumor benefit. This research presents a promising therapeutic approach for DOX-induced CICI, enhancing the safety and efficacy of cancer treatments.


Asunto(s)
Antagonistas de los Receptores CCR5 , Doxorrubicina , Maraviroc , Ratones Endogámicos C57BL , FN-kappa B , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal , Animales , Maraviroc/farmacología , Antagonistas de los Receptores CCR5/farmacología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Femenino , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/inducido químicamente , Neoplasias de la Mama/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Antibióticos Antineoplásicos/toxicidad , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Receptores CCR5/metabolismo , Fármacos Neuroprotectores/farmacología , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/metabolismo
5.
Cancer Res ; 84(13): 2041-2042, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38657103

RESUMEN

Many patients with cancer are at risk of developing cognitive symptoms that often become evident during or after cancer-directed therapy and may have difficulties with attention, concentration, multitasking, executive function, and memory. Despite recent advances in identifying potential molecular and cellular mechanisms underlying cancer and chemotherapy-related cognitive impairment, there is generally a lack of effective treatment strategies, and the development of novel therapeutic interventions represents a major unmet medical need in clinical practice. A recent study by Kim and colleagues suggests that multisensory 40-Hz gamma entrainment using sensory stimuli with combined visual and auditory stimuli is associated with powerful neuroprotective effects in mouse models of cisplatin- or methotrexate-induced "chemobrain." Although the study has some limitations and successful interventions in animal models have often failed to translate into clinical practice, this noninvasive treatment modality has shown promise in preserving brain structure and function and could be tested in patients with cancer who are at risk of cognitive decline.


Asunto(s)
Deterioro Cognitivo Relacionado con la Quimioterapia , Humanos , Animales , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Antineoplásicos/efectos adversos , Ratones
6.
Toxicol Appl Pharmacol ; 485: 116875, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38437957

RESUMEN

Cisplatin is an effective and commonly used chemotherapeutic drug; however, its use is accompanied by several adverse effects, including chemobrain. Ondansetron is a 5-HT3 antagonist, commonly used in prophylactic against chemotherapy-induced nausea and vomiting. Moreover, it has been identified as a novel neuroprotective agent in different animal models. However, its protective role against chemotherapy-induced chemobrain has not been investigated. The current study was the first study that explored the potential neuroprotective effect of ondansetron against cisplatin-induced chemobrain in rats. Cisplatin (5 mg/Kg) was injected intraperitoneally, once weekly, for 4 weeks with the daily administration of ondansetron (0.5 and 1 mg/Kg). Compared to the cisplatin-treated group, ondansetron administration showed a significant decrease in the latency time and a significant increase in ambulation, rearing, and grooming frequency in the open field test (OFT). Moreover, a significant improvement in the latency time in the rotarod and passive avoidance tests, following ondansetron administration. In addition, ondansetron treatment increased the percentage of alternation in the Y-maze test. Also, ondansetron showed a remarkable enhancement in the biochemical parameters in the hippocampus. It increased the acetylcholine (Ach) level and decreased the level of the acetylcholine esterase enzyme (AchE). Ondansetron significantly decreased interleukin-1ß (Il-1ß), tumor necrosis factor-alpha (TNF-α), toll-like receptor-4 (TLR-4), NOD-like receptor-3 (NLRP3) inflammasome as well as caspase-1 and caspase-3 levels. Furthermore, ondansetron significantly decreased the levels of copper transporter-1(CTR1) expression in the hippocampus. Collectively, these findings suggest that ondansetron may exhibit a neuroprotective and therapeutic activity against cisplatin-induced chemobrain.


Asunto(s)
Conducta Animal , Cisplatino , Inflamasomas , Ondansetrón , Animales , Ondansetrón/farmacología , Cisplatino/toxicidad , Masculino , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Ratas , Regulación hacia Abajo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratas Wistar , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Antineoplásicos/toxicidad , Transducción de Señal/efectos de los fármacos , Antagonistas del Receptor de Serotonina 5-HT3/farmacología , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico
7.
Neurosci Biobehav Rev ; 148: 105120, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36906244

RESUMEN

Chemotherapy-induced cognitive impairment (CICI) is a debilitating condition resulting from chemotherapy administration for cancer treatment. CICI is characterised by various cognitive impairments, including issues with learning, memory, and concentration, impacting quality of life. Several neural mechanisms are proposed to drive CICI, including inflammation, therefore, anti-inflammatory agents could ameliorate such impairments. Research is still in the preclinical stage; however, the efficacy of anti-inflammatories to reduce CICI in animal models is unknown. Therefore, a systematic review was conducted, with searches performed in PubMed, Scopus, Embase, PsycInfo and Cochrane Library. A total of 64 studies were included, and of the 50 agents identified, 41 (82%) reduced CICI. Interestingly, while non-traditional anti-inflammatory agents and natural compounds reduced impairment, the traditional agents were unsuccessful. Such results must be taken with caution due to the heterogeneity observed in terms of methods employed. Nevertheless, preliminary evidence suggests anti-inflammatory agents could be beneficial for treating CICI, although it may be critical to think beyond the use of traditional anti-inflammatories when considering which specific compounds to prioritise in development.


Asunto(s)
Antineoplásicos , Deterioro Cognitivo Relacionado con la Quimioterapia , Disfunción Cognitiva , Animales , Antineoplásicos/efectos adversos , Calidad de Vida , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/tratamiento farmacológico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico
8.
J Control Release ; 355: 259-272, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36739910

RESUMEN

The study's primary goal was to enhance medicinal potential of piperine (PIP)-loaded zeolitic imidazolate frameworks-8 (PIP@ZIF-8) against doxorubicin (DOX)-induced cognitive impairments in zebrafish. Herein, PIP@ZIF-8 was synthesized via easy, economical and reproducible ultrasonication method followed by spray drying technology. ZIF-8's structural integrity has been confirmed by PXRD, and even after PIP was encapsulated, the structure of ZIF-8 remained unchanged. Pure ZIF-8 and PIP@ZIF-8 were subjected to TEM analysis, which revealed hexagonal morphology with a nanosize range. FTIR and UV-Visible spectroscopy studies confirmed the drug loading of ZIF-8. Studies on in vitro release revealed 71.48 ± 7.21% and 34.56 ± 5.35% PIP release from PIP@ZIF-8 and unformulated PIP, respectively in pH 7.4. The highest antioxidant scavenging results were obtained with vitamin C (73.77 ± 6.7%) at an intensity of 200 µg/ml, though it was 65.09 ± 2.5% and 57.99 ± 3.1% for PIP@ZIF-8 and PIP, respectively. In vivo studies on zebrafish showed that DOX administration remarkably impaired cognitive activity in T-Maze, and downregulated spatial memory and locomotor activity in the open field test. In addition, DOX administration caused a downregulation in GSH and SOD levels and increase in LPO, AChE and TNF-α levels compared to the vehicle group along with changes in brain histopathology. Further, PIP@ZIF-8 reversed the DOX-induced cognitive impairments by its antioxidant and neuroprotective properties. It can be concluded that PIP@ZIF-8 has a promising therapeutic potential against the chemotherapy-induced cognitive impairments in zebrafish.


Asunto(s)
Deterioro Cognitivo Relacionado con la Quimioterapia , Estructuras Metalorgánicas , Animales , Estructuras Metalorgánicas/química , Pez Cebra , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Antioxidantes , Doxorrubicina/química
9.
Neurosci Lett ; 793: 136975, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36427814

RESUMEN

Chemotherapy results in long-term effects on cognitive dysfunction called chemotherapy-induced cognitive impairment (CICI) in cancer survivors. However, little is known about the potential molecular mechanisms of CICI. This study aimed to determine the role and potential underlying mechanisms of the nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome in cognitive impairments induced by chemotherapeutic agents commonly used in breast cancer. The cognitive effects of chemotherapy were investigated in a rat model using the cocktail of doxorubicin and cyclophosphamide. The NLRP3 pathway was found to be differentially expressed after chemotherapy by iTRAQ-based proteomic analysis of normal and chemotherapeutic hippocampi. Treatment with the NLRP3 inhibitor MCC950 following chemotherapy significantly reduced cognitive impairment and decreased the expression of NLRP3, caspase-1 and ASC. Chemotherapy led to increased expression of the glial response markers Iba-1 and GFAP and the axonal injury markers NF-L and NF-M, an elevated number of apoptotic cells and enhanced microstructural damage to axons and mitochondria, while MCC950 treatment alleviated the glial response, cell death and axonal injury. The protective effect of MCC950 was related to the NLRP3 pathway and levels of inflammatory cytokines (TNF-α, IL-1ß, IL-18, IL-6, IL-4, and IL-10) and oxidative stress-responsive markers (SOD, MDA, CAT and GSH). The results indicate that CICI is associated with NLRP3 pathway-induced oxidative damage and the inflammatory response and provide a potential therapeutic target to treat cognitive impairment after chemotherapy (doxorubicin and cyclophosphamide).


Asunto(s)
Deterioro Cognitivo Relacionado con la Quimioterapia , Disfunción Cognitiva , Indenos , Animales , Ratas , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteómica , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Inflamasomas/metabolismo , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/metabolismo , Furanos/farmacología , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Doxorrubicina/toxicidad , Ciclofosfamida
10.
FEBS J ; 289(20): 6435-6455, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35514149

RESUMEN

Doxorubicin (DOX), an effective, extensively used chemotherapeutic drug, can cause cognitive deterioration in cancer patients. The associated debilitating neurological sequelae are referred to as chemobrain. Our recent work demonstrated that Dox treatment resulted in an imbalance in mitochondrial dynamics, ultimately culminating in cognitive decline in rats. Therefore, in this study, we aim to explore the therapeutic efficacy of a pharmacological intervention, which modulates mitochondrial dynamics using a potent mitochondrial fission inhibitor (Mdivi-1) and mitochondrial fusion promoter (M1) against Dox-induced chemobrain. In the study, male Wistar rats were randomly assigned to receive either normal saline solution or six doses of Dox (3 mg·kg-1 ) via intraperitoneal injection. Then, the Dox-treated rats were intraperitoneally given either 1% DMSO as the vehicle, Mdivi-1 (1.2 mg·kg-1 ), M1 (2 mg·kg-1 ), or a combined treatment of Mdivi-1 and M1 for 30 consecutive days. Long-term learning and memory were evaluated using the novel object location task and novel object recognition task. Following euthanasia, the rat brains were dissected to enable further molecular investigation. We demonstrated that long-term treatment with mitochondrial dynamic modulators suppressed mitochondrial fission in the hippocampus following Dox treatment, leading to an improvement in brain homeostasis. Mitochondrial dynamic modulator treatments restored cognitive function in Dox-treated rats by attenuating neuroinflammation, decreasing oxidative stress, preserving synaptic integrity, reducing potential Alzheimer's related lesions, and mitigating both apoptosis and necroptosis following Dox administration. Together, our findings suggested that mitochondrial dynamics modulators protected against Dox-induced cognitive impairment by rebalancing mitochondrial homeostasis and attenuating both oxidative and inflammatory insults.


Asunto(s)
Deterioro Cognitivo Relacionado con la Quimioterapia , Dinámicas Mitocondriales , Animales , Apoptosis , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Deterioro Cognitivo Relacionado con la Quimioterapia/genética , Cognición , Dimetilsulfóxido/farmacología , Doxorrubicina/efectos adversos , Masculino , Mitocondrias/patología , Enfermedades Neuroinflamatorias , Estrés Oxidativo , Ratas , Ratas Wistar , Solución Salina/farmacología
11.
Int J Mol Sci ; 23(4)2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-35216124

RESUMEN

Advances in the early diagnosis and treatment have led to increases in breast cancer survivorship. Survivors report cognitive impairment symptoms such as loss of concentration and learning and memory deficits which significantly reduce the patient's quality of life. Additional therapies are needed to prevent these side effects and, the precise mechanisms of action responsible are not fully elucidated. However, increasing evidence points toward the use of neuroprotective compounds with antioxidants and anti-inflammatory properties as tools for conserving learning and memory. Here, we examine the ability of piperlongumine (PL), an alkaloid known to have anti-inflammatory and antioxidant effects, to play a neuroprotective role in 16-week-old female C57BL/6J mice treated with a common breast cancer regimen of doxorubicin, cyclophosphamide, and docetaxel (TAC). During social memory testing, TAC-treated mice exhibited impairment, while TAC/PL co-treated mice did not exhibit measurable social memory deficits. Proteomics analysis showed ERK1/2 signaling is involved in TAC and TAC/PL co-treatment. Reduced Nrf2 mRNA expression was also observed. mRNA levels of Gria2 were increased in TAC treated mice and reduced in TAC/PL co-treated mice. In this study, PL protects against social memory impairment when co-administered with TAC via multifactorial mechanisms involving oxidative stress and synaptic plasticity.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Disfunción Cognitiva/inducido químicamente , Disfunción Cognitiva/tratamiento farmacológico , Dioxolanos/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antioxidantes/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Deterioro Cognitivo Relacionado con la Quimioterapia/metabolismo , Disfunción Cognitiva/metabolismo , Femenino , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Calidad de Vida , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos
12.
Acta Neuropathol Commun ; 10(1): 11, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-35093182

RESUMEN

Cisplatin is used to combat solid tumors. However, patients treated with cisplatin often develop cognitive impairments, sensorimotor deficits, and peripheral neuropathy. There is no FDA-approved treatment for these neurotoxicities. We investigated the capacity of a highly selective A3 adenosine receptor (AR) subtype (A3AR) agonist, MRS5980, to prevent and reverse cisplatin-induced neurotoxicities. MRS5980 prevented cisplatin-induced cognitive impairment (decreased executive function and impaired spatial and working memory), sensorimotor deficits, and neuropathic pain (mechanical allodynia and spontaneous pain) in both sexes. At the structural level, MRS5980 prevented the cisplatin-induced reduction in markers of synaptic integrity. In-situ hybridization detected Adora3 mRNA in neurons, microglia, astrocytes and oligodendrocytes. RNAseq analysis identified 164 genes, including genes related to mitochondrial function, of which expression was changed by cisplatin and normalized by MRS5980. Consistently, MRS5980 prevented cisplatin-induced mitochondrial dysfunction and decreased signs of oxidative stress. Transcriptomic analysis showed that the A3AR agonist upregulates genes related to repair pathways including NOTCH1 signaling and chromatin modification in the cortex of cisplatin-treated mice. Importantly, A3AR agonist administration after completion of cisplatin treatment resolved cognitive impairment, neuropathy and sensorimotor deficits. Our results highlight the efficacy of a selective A3AR agonist to prevent and reverse cisplatin-induced neurotoxicities via preventing brain mitochondrial damage and activating repair pathways. An A3AR agonist is already in cancer, clinical trials and our results demonstrate management of neurotoxic side effects of chemotherapy as an additional therapeutic benefit.


Asunto(s)
Agonistas del Receptor de Adenosina A3/farmacología , Antineoplásicos/efectos adversos , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Cisplatino/efectos adversos , Receptor de Adenosina A3/metabolismo , Memoria Espacial/efectos de los fármacos , Agonistas del Receptor de Adenosina A3/uso terapéutico , Animales , Femenino , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Dolor/metabolismo
13.
Eur J Cancer ; 161: 10-22, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34896904

RESUMEN

BACKGROUND: Chemotherapy-related cognitive impairment (CRCI) is highly prevalent in patients with cancer and is associated with poor outcomes and quality of life. To date, the management of CRCI remains a clinical challenge. Herein, we aim to determine the preventive effects of probiotics on CRCI development and underlying mechanisms. METHODS: We conducted a randomised, double-blind and placebo-controlled trial (ChiCTR-INQ-17014181) of 159 patients with breast cancer and further investigated the underlying mechanism in a pre-clinical setting. From 2018 to 2019, patients with breast cancer (Stage I-III) who needed adjuvant chemotherapy were screened, enrolled and randomly assigned to receive either probiotics or placebo (three capsules, twice/day) during chemotherapy. Their cognition, anxiety and depression were assessed with well-established assays; their plasma biomarkers, metabolites and faecal microbiota compositions were measured. In addition, the systemic effects of the metabolites found in the clinical trial on long-term potentiation, synapse injury, oxidative stress and glial activation were assessed in rats. RESULTS: Probiotics supplement significantly decreased the incidence of CRCI, improved the allover cognitive functions, changed the gut microbial composition and modulated nine plasma metabolite changes. Among these metabolites, p-Mentha-1,8-dien-7-ol, Linoelaidyl carnitine and 1-aminocyclopropane-1-carboxylic acid were negatively correlated with the occurrence of CRCI. Furthermore, probiotics supplement increased plasma p-Mentha-1,8-dien-7-ol in rats. Administration of exogenous p-Mentha-1,8-dien-7-ol significantly alleviated chemotherapy-induced long-term potentiation impairment, synapse injury, oxidative stress and glial activation in the hippocampus of rats. CONCLUSION: Our data indicated that probiotics supplement prevents the occurrence of CRCI in patients with breast cancer via modulating plasma metabolites, including p-Mentha-1,8-dien-7-ol. TRIAL REGISTRATION: Chinese Clinical Trial Registry (ChiCTR-INQ-17014181) [http://www.chictr.org.cn/showproj.aspx?proj=24294].


Asunto(s)
Neoplasias de la Mama/complicaciones , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Probióticos/uso terapéutico , Adulto , Método Doble Ciego , Femenino , Humanos , Persona de Mediana Edad , Probióticos/farmacología , Estudios Prospectivos , Adulto Joven
14.
Neurotoxicology ; 88: 1-13, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34656704

RESUMEN

Cognitive impairment or "chemobrain" is a troublesome adverse effect which had been increasingly reported by cancer patients after doxorubicin (DOX) chemotherapy. Notably, Hypertension, a very common comorbidity in cancer patients, could pose a greater risk for negative cognitive outcomes. Amiloride (AML) is an antihypertensive, potassium-sparing diuretic that has been proven to be neuroprotective in different experimental models; this can be attributed to its ability to inhibit different ion transporters such as Na+/H+ exchanger (NHE), which upon excessive activation can result in intracellular cationic overload, followed by oxidative damage and cellular death. Accordingly, this study was designed to investigate the potential neuroprotective effect of AML against DOX-induced chemobrain and to elucidate possible underlying mechanisms. Briefly, Histopathological examination and neurobehavioral testing (Morris water maze, Y maze and passive avoidance test) showed that AML co-treatment (10 mg/kg/day) markedly attenuated DOX (2 mg/kg/week)-induced neurodegeneration and memory impairment after 4 weeks of treatments. We found that DOX administration up-regulated NHE expression and increased lactic acid content in the hippocampus which were markedly opposed by AML. Moreover, AML mitigated DOX-induced neuroinflammation and decreased hippocampal tumor necrosis factor-α level, nuclear factor kappa-B, and cyclooxygenase-2 expression. Additionally, AML counteracted DOX-induced hippocampal oxidative stress as indicated by normalized malondialdehyde and glutathione levels. Furthermore, AML halted DOX-induced hippocampal apoptosis as evidenced by decreased caspase-3 activity and lower cytochrome c immunoexpression. Our results in addition to the previously reported antitumor effects of AML and its ability to mitigate cancer resistance to DOX therapy could point toward possible new repositioning scenarios of the diuretic AML especially regarding hypertensive cancer patients.


Asunto(s)
Amilorida/farmacología , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Diuréticos/farmacología , Doxorrubicina/toxicidad , Animales , Deterioro Cognitivo Relacionado con la Quimioterapia/etiología , Doxorrubicina/antagonistas & inhibidores , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Enfermedades Neuroinflamatorias/inducido químicamente , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Intercambiadores de Sodio-Hidrógeno/metabolismo
15.
Int J Mol Sci ; 22(23)2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34884513

RESUMEN

A wide range of cognitive deficits, including memory loss associated with hippocampal dysfunction, have been widely reported in cancer survivors who received chemotherapy. Changes in both white matter and gray matter volume have been observed following chemotherapy treatment, with reduced volume in the medial temporal lobe thought to be due in part to reductions in hippocampal neurogenesis. Pre-clinical rodent models confirm that common chemotherapeutic agents used to treat various forms of non-CNS cancers reduce rates of hippocampal neurogenesis and impair performance on hippocampally-mediated learning and memory tasks. We review the pre-clinical rodent literature to identify how various chemotherapeutic drugs affect hippocampal neurogenesis and induce cognitive impairment. We also review factors such as physical exercise and environmental stimulation that may protect against chemotherapy-induced neurogenic suppression and hippocampal neurotoxicity. Finally, we review pharmacological interventions that target the hippocampus and are designed to prevent or reduce the cognitive and neurotoxic side effects of chemotherapy.


Asunto(s)
Antineoplásicos/efectos adversos , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Animales , Deterioro Cognitivo Relacionado con la Quimioterapia/etiología , Deterioro Cognitivo Relacionado con la Quimioterapia/patología , Humanos , Neoplasias/patología
16.
PLoS One ; 16(6): e0252522, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34081735

RESUMEN

While chemotherapy is the most effective therapeutic approach for treating a variety of cancer patients, commonly used chemotherapeutic agents, often induce several adverse effects. Escalating evidence indicates that chemotherapeutics, particularly doxorubicin (DOX) and cyclophosphamide (CPS), induce cognitive impairment associated with central nervous system toxicity. This study was performed to determine neuroprotective effects of Oroxylum indicum extract (OIE) in regard to preventing chemotherapy induced cognitive impairment (CICI) occurring after 4 cycles of DOX (2mg/kg) and CPS (50mg/kg) combination chemotherapy in male C57BL/6J mice. OIE significantly prevented the chemotherapy impaired short-term cognitive performance, exploratory behavior associated with cognitive performance, cognitive performance, and spatial learning and memory in the Y-maze, Open-Field, Novel Object Recognition, and Morris Water Maze tests, respectively. These data suggest that OIE protects from the CICI. OIE decreased the reactive oxygen species and lipid peroxide generated by the chemotherapy treatment in the brain, while also blocking the chemotherapy-induced glutathione depletion. These results establish that OIE exhibits potent antioxidant activity in chemotherapy treated mice. Notably, OIE significantly increased the Complex-I and Complex-IV activities in the brain, indicating that OIE enhances mitochondrial function in the brain. In silico analysis of the major active chemical constituents (Oroxylin A, Baicalein and Chrysin) of OIE indicated that OIE has a favorable absorption, distribution, metabolism and excretion (ADME) profile. Taken together, our results are consistent with the conclusion that OIE prevents CICI by counteracting oxidative stress and perhaps by improving mitochondrial function.


Asunto(s)
Encéfalo/metabolismo , Deterioro Cognitivo Relacionado con la Quimioterapia/fisiopatología , Disfunción Cognitiva/fisiopatología , Animales , Antineoplásicos/uso terapéutico , Encéfalo/efectos de los fármacos , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Disfunción Cognitiva/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/uso terapéutico
17.
Curr Treat Options Oncol ; 22(8): 67, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34110516

RESUMEN

OPINION STATEMENT: One of the most burdensome symptoms reported by breast cancer patients is chemotherapy-related neurocognitive impairment. It is estimated that of the 11 million cancer survivors in the USA, 22% of them are breast cancer patients. The National Cancer Institute classified chemotherapy-related cognitive impairment (CRCI) as one of the most debilitating sequelae of cancer therapy, limiting this patient population from recommencing their lives prior to the diagnosis of breast cancer. Currently, there are no strategies that are established to prevent, mitigate, or treat CRCI. In addition to surviving cancer, quality of life is critical to cancer survivors. Based on the multiple and complex biological and psychosocial etiology, the varying manifestation and extent of cognitive decline documented in breast cancer survivors, possibly attributed to varying combinations of chemotherapy and dose and duration of therapy, multimodal interventions combining promising nutrient-derived bioactive compounds with antioxidant and anti-inflammatory properties, in addition to structured cognitive training and exercise regimens, can work synergistically to reduce inflammation and oxidative stress with significant improvement in cognitive function resulting in improvements in quality of life of breast cancer survivors.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Supervivientes de Cáncer , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Neoplasias de la Mama/complicaciones , Deterioro Cognitivo Relacionado con la Quimioterapia/etiología , Dieta , Femenino , Humanos , Nutrientes
19.
Life Sci ; 269: 119078, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33460662

RESUMEN

AIMS: Cognitive decline is one of the most challenging issues for cancer survivors undergoing doxorubicin (DOX) based chemotherapy. Oxidative stress and inflammation primarily through tumor necrosis factor-alpha (TNF-α) are considered the key contributors to DOX-induced chemobrain. Berberine (BBR) has attracted much interest because of its anti-oxidative, anti-inflammatory and anti-apoptotic actions. This study aimed to evaluate the potential neuroprotective effect of BBR in DOX-induced neurodegeneration and cognitive deficits. MATERIALS AND METHODS: Chemobrain was induced by DOX i.p. injection at the dose of 2 mg/kg, once/week, for four consecutive weeks. Rats were treated with BBR (100 mg/kg, p.o.) for 5 days/week for four consecutive weeks. KEY FINDINGS: BBR significantly attenuated behavioral defects in DOX-induced cognitive impairment. Besides, BBR reversed histopathological abnormalities. Mechanistically, it reversed DOX-induced neuroinflammation by attenuating NF-κB gene and protein expression in addition to diminishing expression of pro-inflammatory mediators (TNF-α and IL-1ß), as well as apoptotic related factors (Bax, Bcl2 and Bax/Bcl2 ratio). Additionally, BBR activated the anti-oxidative defense via upregulating the expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and manganese superoxide dismutase (MnSOD). BBR improved synaptic plasticity through cAMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF). These effects were related through the modulation of Sirtuin1 (SIRT1) expression. SIGNIFICANCE: BBR is highlighted to induce neuroprotection against DOX-induced cognitive decline through modulating brain growth factors and imposing an anti-inflammatory, anti-apoptotic and anti-oxidative effects.


Asunto(s)
Conducta Animal/efectos de los fármacos , Berberina/farmacología , Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Doxorrubicina/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/toxicidad , Deterioro Cognitivo Relacionado con la Quimioterapia/etiología , Deterioro Cognitivo Relacionado con la Quimioterapia/metabolismo , Deterioro Cognitivo Relacionado con la Quimioterapia/patología , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Masculino , Ratas , Transducción de Señal
20.
Neurochem Res ; 46(2): 149-158, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33237471

RESUMEN

Chemotherapy-induced cognitive impairment (CICI) is a common detrimental effect of cancer treatment, occurring in up to 75% of cancer patients. The widely utilized chemotherapeutic agent doxorubicin (DOX) has been implicated in cognitive decline, mostly via cytokine-induced neuroinflammatory and oxidative and mitochondrial damage to brain tissues. C-phycocyanin (CP) has previously been shown to have potent anti-inflammatory, antioxidant, and mitochondrial protective properties. Therefore, this present study was aimed to investigate the neuroprotective effects of CP against DOX-elicited cognitive impairment and explore the underlying mechanisms. CP treatment (50 mg/kg) significantly improved behavioral deficits in DOX-treated mice. Furthermore, CP suppressed DOX-induced neuroinflammation and oxidative stress, mitigated mitochondrial abnormalities, rescued dendritic spine loss, and increased synaptic density in the hippocampus of DOX-treated mice. Our results suggested that CP improves established DOX-induced cognitive deficits, which could be explained at least partly by inhibition of neuroinflammatory and oxidant stress and attenuation of mitochondrial and synaptic dysfunction.


Asunto(s)
Deterioro Cognitivo Relacionado con la Quimioterapia/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Ficocianina/uso terapéutico , Sinapsis/efectos de los fármacos , Animales , Deterioro Cognitivo Relacionado con la Quimioterapia/complicaciones , Deterioro Cognitivo Relacionado con la Quimioterapia/patología , Espinas Dendríticas/efectos de los fármacos , Doxorrubicina , Hipocampo/efectos de los fármacos , Hipocampo/patología , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Prueba del Laberinto Acuático de Morris/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...