98 results on '"Ines Moreno-Gonzalez"'
Search Results
2. Monocyte-derived cells invade brain parenchyma and amyloid plaques in human Alzheimer’s disease hippocampus
- Author
-
Clara Muñoz-Castro, Marina Mejias-Ortega, Elisabeth Sanchez-Mejias, Victoria Navarro, Laura Trujillo-Estrada, Sebastian Jimenez, Juan Antonio Garcia-Leon, Juan Jose Fernandez-Valenzuela, Maria Virtudes Sanchez-Mico, Carmen Romero-Molina, Ines Moreno-Gonzalez, David Baglietto-Vargas, Marisa Vizuete, Antonia Gutierrez, and Javier Vitorica
- Subjects
Alzheimer’s disease ,Myeloid cells ,Microglia ,Brain infiltration ,Human hippocampus ,Amyloid plaques ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Microglia are brain-resident myeloid cells and play a major role in the innate immune responses of the CNS and the pathogenesis of Alzheimer's disease (AD). However, the contribution of nonparenchymal or brain-infiltrated myeloid cells to disease progression remains to be demonstrated. Here, we show that monocyte-derived cells (MDC) invade brain parenchyma in advanced stages of AD continuum using transcriptional analysis and immunohistochemical characterization in post-mortem human hippocampus. Our findings demonstrated that a high proportion (60%) of demented Braak V–VI individuals was associated with up-regulation of genes rarely expressed by microglial cells and abundant in monocytes, among which stands the membrane-bound scavenger receptor for haptoglobin/hemoglobin complexes or Cd163. These Cd163-positive MDC invaded the hippocampal parenchyma, acquired a microglial-like morphology, and were located in close proximity to blood vessels. Moreover, and most interesting, these invading monocytes infiltrated the nearby amyloid plaques contributing to plaque-associated myeloid cell heterogeneity. However, in aged-matched control individuals with hippocampal amyloid pathology, no signs of MDC brain infiltration or plaque invasion were found. The previously reported microglial degeneration/dysfunction in AD hippocampus could be a key pathological factor inducing MDC recruitment. Our data suggest a clear association between MDC infiltration and endothelial activation which in turn may contribute to damage of the blood brain barrier integrity. The recruitment of monocytes could be a consequence rather than the cause of the severity of the disease. Whether monocyte infiltration is beneficial or detrimental to AD pathology remains to be fully elucidated. These findings open the opportunity to design targeted therapies, not only for microglia but also for the peripheral immune cell population to modulate amyloid pathology and provide a better understanding of the immunological mechanisms underlying the progression of AD.
- Published
- 2023
- Full Text
- View/download PDF
3. Aβ oligomers trigger necroptosis-mediated neurodegeneration via microglia activation in Alzheimer’s disease
- Author
-
Natalia Salvadores, Ines Moreno-Gonzalez, Nazaret Gamez, Gabriel Quiroz, Laura Vegas-Gomez, Marcela Escandón, Sebastian Jimenez, Javier Vitorica, Antonia Gutierrez, Claudio Soto, and Felipe A. Court
- Subjects
Alzheimer’s disease ,Amyloid-β oligomers ,Necroptosis ,Microglia ,Neurodegeneration ,Neuroprotection ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Alzheimer’s disease (AD) is a major adult-onset neurodegenerative condition with no available treatment. Compelling reports point amyloid-β (Aβ) as the main etiologic agent that triggers AD. Although there is extensive evidence of detrimental crosstalk between Aβ and microglia that contributes to neuroinflammation in AD, the exact mechanism leading to neuron death remains unknown. Using postmortem human AD brain tissue, we show that Aβ pathology is associated with the necroptosis effector pMLKL. Moreover, we found that the burden of Aβ oligomers (Aβo) correlates with the expression of key markers of necroptosis activation. Additionally, inhibition of necroptosis by pharmacological or genetic means, reduce neurodegeneration and memory impairment triggered by Aβo in mice. Since microglial activation is emerging as a central driver for AD pathogenesis, we then tested the contribution of microglia to the mechanism of Aβo-mediated necroptosis activation in neurons. Using an in vitro model, we show that conditioned medium from Aβo-stimulated microglia elicited necroptosis in neurons through activation of TNF-α signaling, triggering extensive neurodegeneration. Notably, necroptosis inhibition provided significant neuronal protection. Together, these findings suggest that Aβo-mediated microglia stimulation in AD contributes to necroptosis activation in neurons and neurodegeneration. As necroptosis is a druggable degenerative mechanism, our findings might have important therapeutic implications to prevent the progression of AD.
- Published
- 2022
- Full Text
- View/download PDF
4. Generation of a humanized Aβ expressing mouse demonstrating aspects of Alzheimer’s disease-like pathology
- Author
-
David Baglietto-Vargas, Stefania Forner, Lena Cai, Alessandra C. Martini, Laura Trujillo-Estrada, Vivek Swarup, Marie Minh Thu Nguyen, Kelly Do Huynh, Dominic I. Javonillo, Kristine Minh Tran, Jimmy Phan, Shan Jiang, Enikö A. Kramár, Cristina Nuñez-Diaz, Gabriela Balderrama-Gutierrez, Franklin Garcia, Jessica Childs, Carlos J. Rodriguez-Ortiz, Juan Antonio Garcia-Leon, Masashi Kitazawa, Mohammad Shahnawaz, Dina P. Matheos, Xinyi Ma, Celia Da Cunha, Ken C. Walls, Rahasson R. Ager, Claudio Soto, Antonia Gutierrez, Ines Moreno-Gonzalez, Ali Mortazavi, Andrea J. Tenner, Grant R. MacGregor, Marcelo Wood, Kim N. Green, and Frank M. LaFerla
- Subjects
Science - Abstract
Most instances of Alzheimer’s disease (AD) are sporadic or not associated with a particular mutation. Here, the authors develop knock-in mice that express wildtype human Aβ under control of the mouse App locus, which may have potential for modelling some aspects of sporadic late onset AD.
- Published
- 2021
- Full Text
- View/download PDF
5. Amyloid pathology arrangements in Alzheimer’s disease brains modulate in vivo seeding capability
- Author
-
Claudia Duran-Aniotz, Ines Moreno-Gonzalez, Nazaret Gamez, Nelson Perez-Urrutia, Laura Vegas-Gomez,, Claudio Soto, and Rodrigo Morales
- Subjects
Alzheimer’s disease ,Amyloid-beta ,Prions ,Strains ,Pathology ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Amyloid-β (Aβ) misfolding is one of the hallmark pathological features of Alzheimer’s disease (AD). AD can manifest with diverse symptomatology including variable rates of cognitive decline, duration of clinical disease, and other detrimental changes. Several reports suggest that conformational diversity in misfolded Aβ is a leading factor for clinical variability in AD, analogous to what it has been described for prion strains in prion diseases. Notably, prion strains generate diverse patterns of misfolded protein deposition in the brains of affected individuals. Here, we tested the in vivo prion-like transmission features of four AD brains displaying particular patterns of amyloidosis. AD brains induced different phenotypes in recipient mice, as evaluated by their specific seeding activity, as well as the total amount of Aβ deposited surrounding vascular structures and the reactivity of amyloid pathology to thioflavin S. Our results support the notion that AD-subtypes are encoded in disease-associated Aβ. Further research exploring whether AD include a spectrum of different clinical conditions or syndromes may pave the way to personalized diagnosis and treatments.
- Published
- 2021
- Full Text
- View/download PDF
6. Editorial: Metabolic Alterations in Neurodegenerative Disorders
- Author
-
Kristine K. Freude, Ines Moreno-Gonzalez, Carlos J. Rodriguez-Ortiz, and David Baglietto-Vargas
- Subjects
neurodegenerative disorders ,hyperlipidemia ,metabolism ,blood-brain barrier ,astrocytes ,ceramides ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2022
- Full Text
- View/download PDF
7. Editorial: Protein Misfolding and Proteostasis Impairment in Aging and Neurodegeneration: From Spreading Studies to Therapeutic Approaches
- Author
-
Claudia Duran-Aniotz, Ines Moreno-Gonzalez, Danilo B. Medinas, and Rodrigo Morales
- Subjects
protein misfolding ,proteostasis ,aging ,neurodegeneration ,Alzheimer's disease ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2022
- Full Text
- View/download PDF
8. G-quadruplexes Stabilization Upregulates CCN1 and Accelerates Aging in Cultured Cerebral Endothelial Cells
- Author
-
Brian Noh, Maria P. Blasco-Conesa, Yun-Ju Lai, Bhanu Priya Ganesh, Akihiko Urayama, Ines Moreno-Gonzalez, Sean P. Marrelli, Louise D. McCullough, and Jose Felix Moruno-Manchon
- Subjects
senescence ,endothelial cells ,G-quadruplex ,aging ,CCN1 ,Geriatrics ,RC952-954.6 - Abstract
Senescence in the cerebral endothelium has been proposed as a mechanism that can drive dysfunction of the cerebral vasculature, which precedes vascular dementia. Cysteine-rich angiogenic inducer 61 (Cyr61/CCN1) is a matricellular protein secreted by cerebral endothelial cells (CEC). CCN1 induces senescence in fibroblasts. However, whether CCN1 contributes to senescence in CEC and how this is regulated requires further study. Aging has been associated with the formation of four-stranded Guanine-quadruplexes (G4s) in G-rich motifs of DNA and RNA. Stabilization of the G4 structures regulates transcription and translation either by upregulation or downregulation depending on the gene target. Previously, we showed that aged mice treated with a G4-stabilizing compound had enhanced senescence-associated (SA) phenotypes in their brains, and these mice exhibited enhanced cognitive deficits. A sequence in the 3′-UTR of the human CCN1 mRNA has the ability to fold into G4s in vitro. We hypothesize that G4 stabilization regulates CCN1 in cultured primary CEC and induces endothelial senescence. We used cerebral microvessel fractions and cultured primary CEC from young (4-months old, m/o) and aged (18-m/o) mice to determine CCN1 levels. SA phenotypes were determined by high-resolution fluorescence microscopy in cultured primary CEC, and we used Thioflavin T to recognize RNA-G4s for fluorescence spectra. We found that cultured CEC from aged mice exhibited enhanced levels of SA phenotypes, and higher levels of CCN1 and G4 stabilization. In cultured CEC, CCN1 induced SA phenotypes, such as SA β-galactosidase activity, and double-strand DNA damage. Furthermore, CCN1 levels were upregulated by a G4 ligand, and a G-rich motif in the 3′-UTR of the Ccn1 mRNA was folded into a G4. In conclusion, we demonstrate that CCN1 can induce senescence in cultured primary CEC, and we provide evidence that G4 stabilization is a novel mechanism regulating the SASP component CCN1.
- Published
- 2022
- Full Text
- View/download PDF
9. Aged Cattle Brain Displays Alzheimer's Disease-Like Pathology and Promotes Brain Amyloidosis in a Transgenic Animal Model
- Author
-
Ines Moreno-Gonzalez, George Edwards, Rodrigo Morales, Claudia Duran-Aniotz, Gabriel Escobedo, Mercedes Marquez, Marti Pumarola, and Claudio Soto
- Subjects
amyloid ,prions ,Alzheimer's disease ,spreading ,protein misfolding ,seeding ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Alzheimer's disease (AD) is one of the leading causes of dementia in late life. Although the cause of AD neurodegenerative changes is not fully understood, extensive evidence suggests that the misfolding, aggregation and cerebral accumulation of amyloid beta (Aβ) and tau proteins are hallmark events. Recent reports have shown that protein misfolding and aggregation can be induced by administration of small quantities of preformed aggregates, following a similar principle by which prion diseases can be transmitted by infection. In the past few years, many of the typical properties that characterize prions as infectious agents were also shown in Aβ aggregates. Interestingly, prion diseases affect not only humans, but also various species of mammals, and it has been demonstrated that infectious prions present in animal tissues, particularly cattle affected by bovine spongiform encephalopathy (BSE), can infect humans. It has been reported that protein deposits resembling Aβ amyloid plaques are present in the brain of several aged non-human mammals, including monkeys, bears, dogs, and cheetahs. In this study, we investigated the presence of Aβ aggregates in the brain of aged cattle, their similarities with the protein deposits observed in AD patients, and their capability to promote AD pathological features when intracerebrally inoculated into transgenic animal models of AD. Our data show that aged cattle can develop AD-like neuropathological abnormalities, including amyloid plaques, as studied histologically. Importantly, cow-derived aggregates accelerate Aβ amyloid deposition in the brain of AD transgenic animals. Surprisingly, the rate of induction produced by administration of the cattle material was substantially higher than induction produced by injection of similar amounts of human AD material. Our findings demonstrate that cows develop seeding-competent Aβ aggregates, similarly as observed in AD patients.
- Published
- 2022
- Full Text
- View/download PDF
10. Plaque-Associated Oligomeric Amyloid-Beta Drives Early Synaptotoxicity in APP/PS1 Mice Hippocampus: Ultrastructural Pathology Analysis
- Author
-
Raquel Sanchez-Varo, Elisabeth Sanchez-Mejias, Juan Jose Fernandez-Valenzuela, Vanessa De Castro, Marina Mejias-Ortega, Angela Gomez-Arboledas, Sebastian Jimenez, Maria Virtudes Sanchez-Mico, Laura Trujillo-Estrada, Ines Moreno-Gonzalez, David Baglietto-Vargas, Marisa Vizuete, Jose Carlos Davila, Javier Vitorica, and Antonia Gutierrez
- Subjects
Alzheimer’s disease ,synaptic pathology ,hippocampus ,transgenic mice (Tg) ,amyloid ,oligomers ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder characterized by initial memory impairments that progress to dementia. In this sense, synaptic dysfunction and loss have been established as the pathological features that best correlate with the typical early cognitive decline in this disease. At the histopathological level, post mortem AD brains typically exhibit intraneuronal neurofibrillary tangles (NFTs) along with the accumulation of amyloid-beta (Abeta) peptides in the form of extracellular deposits. Specifically, the oligomeric soluble forms of Abeta are considered the most synaptotoxic species. In addition, neuritic plaques are Abeta deposits surrounded by activated microglia and astroglia cells together with abnormal swellings of neuronal processes named dystrophic neurites. These periplaque aberrant neurites are mostly presynaptic elements and represent the first pathological indicator of synaptic dysfunction. In terms of losing synaptic proteins, the hippocampus is one of the brain regions most affected in AD patients. In this work, we report an early decline in spatial memory, along with hippocampal synaptic changes, in an amyloidogenic APP/PS1 transgenic model. Quantitative electron microscopy revealed a spatial synaptotoxic pattern around neuritic plaques with significant loss of periplaque synaptic terminals, showing rising synapse loss close to the border, especially in larger plaques. Moreover, dystrophic presynapses were filled with autophagic vesicles in detriment of the presynaptic vesicular density, probably interfering with synaptic function at very early synaptopathological disease stages. Electron immunogold labeling showed that the periphery of amyloid plaques, and the associated dystrophic neurites, was enriched in Abeta oligomers supporting an extracellular location of the synaptotoxins. Finally, the incubation of primary neurons with soluble fractions derived from 6-month-old APP/PS1 hippocampus induced significant loss of synaptic proteins, but not neuronal death. Indeed, this preclinical transgenic model could serve to investigate therapies targeted at initial stages of synaptic dysfunction relevant to the prodromal and early AD.
- Published
- 2021
- Full Text
- View/download PDF
11. Editorial: Risk Factors for Alzheimer's Disease
- Author
-
Ines Moreno-Gonzalez, Rodrigo Morales, David Baglietto-Vargas, and Raquel Sanchez-Varo
- Subjects
Alzheimer's disease ,risk factors ,modifiable ,genetic variance ,peripheral ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2020
- Full Text
- View/download PDF
12. Longitudinal Assessment of Tau-Associated Pathology by 18F-THK5351 PET Imaging: A Histological, Biochemical, and Behavioral Study
- Author
-
Ines Moreno-Gonzalez, George A. Edwards, Omar Hasan, Nazaret Gamez, Jonathan E. Schulz, Juan Jose Fernandez-Valenzuela, Antonia Gutierrez, Claudio Soto, and Paul E. Schulz
- Subjects
PET imaging ,Tau ,THK5351 ,tracer ,biomarker ,histological assessment ,Medicine (General) ,R5-920 - Abstract
Several common and debilitating neurodegenerative disorders are characterized by the intracellular accumulation of neurofibrillary tangles (NFTs), which are composed of hyperphosphorylated tau protein. In Alzheimer’s disease (AD), NFTs are accompanied by extracellular amyloid-beta (Aβ), but primary tauopathy disorders are marked by the accumulation of tau protein alone, including forms of frontotemporal dementia (FTD), corticobasal degeneration (CBD), and progressive supranuclear palsy (PSP), among others. 18F-THK5351 has been reported to bind pathological tau as well as associated reactive astrogliosis. The goal of this study was to validate the ability of the PET tracer 18F-THK5351 to detect early changes in tau-related pathology and its relation to other pathological hallmarks. We demonstrated elevated in vivo 18F-THK5351 PET signaling over time in transgenic P301S tau mice from 8 months that had a positive correlation with histological and biochemical tau changes, as well as motor, memory, and learning impairment. This study indicates that 18F-THK5351 may help fill a critical need to develop PET imaging tracers that detect aberrant tau aggregation and related neuropathology in order to diagnose the onset of tauopathies, gain insights into their underlying pathophysiologies, and to have a reliable biomarker to follow during treatment trials.
- Published
- 2021
- Full Text
- View/download PDF
13. Inflammatory Cascade in Alzheimer’s Disease Pathogenesis: A Review of Experimental Findings
- Author
-
Jade de Oliveira, Ewa Kucharska, Michelle Lima Garcez, Matheus Scarpatto Rodrigues, João Quevedo, Ines Moreno-Gonzalez, and Josiane Budni
- Subjects
Alzheimer’s disease ,neurodegenerative disease ,dementia ,neuroinflammation ,inflammatory cascade ,systemic inflammation ,Cytology ,QH573-671 - Abstract
Alzheimer’s disease (AD) is the leading cause of dementia worldwide. Most AD patients develop the disease in late life, named late onset AD (LOAD). Currently, the most recognized explanation for AD pathology is the amyloid cascade hypothesis. It is assumed that amyloid beta (Aβ) aggregation and deposition are critical pathogenic processes in AD, leading to the formation of amyloid plaques, as well as neurofibrillary tangles, neuronal cell death, synaptic degeneration, and dementia. In LOAD, the causes of Aβ accumulation and neuronal loss are not completely clear. Importantly, the blood–brain barrier (BBB) disruption seems to present an essential role in the induction of neuroinflammation and consequent AD development. In addition, we propose that the systemic inflammation triggered by conditions like metabolic diseases or infections are causative factors of BBB disruption, coexistent inflammatory cascade and, ultimately, the neurodegeneration observed in AD. In this regard, the use of anti-inflammatory molecules could be an interesting strategy to treat, delay or even halt AD onset and progression. Herein, we review the inflammatory cascade and underlying mechanisms involved in AD pathogenesis and revise the anti-inflammatory effects of compounds as emerging therapeutic drugs against AD.
- Published
- 2021
- Full Text
- View/download PDF
14. Modifiable Risk Factors for Alzheimer’s Disease
- Author
-
George A. Edwards III, Nazaret Gamez, Gabriel Escobedo Jr., Olivia Calderon, and Ines Moreno-Gonzalez
- Subjects
Alzheimer’s disease ,risk factors ,comorbidities ,vascular disease ,traumatic brain injury ,epilepsy ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Since first described in the early 1900s, Alzheimer’s disease (AD) has risen exponentially in prevalence and concern. Research still drives to understand the etiology and pathogenesis of this disease and what risk factors can attribute to AD. With a majority of AD cases being of sporadic origin, the increasing exponential growth of an aged population and a lack of treatment, it is imperative to discover an easy accessible preventative method for AD. Some risk factors can increase the propensity of AD such as aging, sex, and genetics. Moreover, there are also modifiable risk factors—in terms of treatable medical conditions and lifestyle choices—that play a role in developing AD. These risk factors have their own biological mechanisms that may contribute to AD etiology and pathological consequences. In this review article, we will discuss modifiable risk factors and discuss the current literature of how each of these factors interplay into AD development and progression and if strategically analyzed and treated, could aid in protection against this neurodegenerative disease.
- Published
- 2019
- Full Text
- View/download PDF
15. Peripheral Delivery of Neural Precursor Cells Ameliorates Parkinson’s Disease-Associated Pathology
- Author
-
George Edwards III, Nazaret Gamez, Enrique Armijo, Carlos Kramm, Rodrigo Morales, Kathleen Taylor-Presse, Paul E. Schulz, Claudio Soto, and Ines Moreno-Gonzalez
- Subjects
neuronal precursors ,stem cells ,parkinson’s disease ,therapy ,intravenous ,inflammation ,clinical symptoms ,Cytology ,QH573-671 - Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disorder characterized by loss of motor control due to a wide loss of dopaminergic neurons along the nigro-striatal pathway. Some of the mechanisms that contribute to this cell death are inflammation, oxidative stress, and misfolded alpha-synuclein-induced toxicity. Current treatments are effective at managing the early motor symptoms of the disease, but they become ineffective over time and lead to adverse effects. Previous research using intracerebral stem cell therapy for treatment of PD has provided promising results; however, this method is very invasive and is often associated with unacceptable side effects. In this study, we used an MPTP-injected mouse model of PD and intravenously administered neural precursors (NPs) obtained from mouse embryonic and mesenchymal stem cells. Clinical signs and neuropathology were assessed. Female mice treated with NPs had improved motor function and reduction in the neuroinflammatory response. In terms of safety, there were no tumorigenic formations or any detectable adverse effect after treatment. Our results suggest that peripheral administration of stem cell-derived NPs may be a promising and safe therapy for the recovery of impaired motor function and amelioration of brain pathology in PD.
- Published
- 2019
- Full Text
- View/download PDF
16. Aggregate-depleted brain fails to induce Aβ deposition in a mouse model of Alzheimer's disease.
- Author
-
Claudia Duran-Aniotz, Rodrigo Morales, Ines Moreno-Gonzalez, Ping Ping Hu, Joseph Fedynyshyn, and Claudio Soto
- Subjects
Medicine ,Science - Abstract
Recent studies in animal models of Alzheimer's disease (AD) show that amyloid-beta (Aβ) misfolding can be transmissible; however, the mechanisms by which this process occurs have not been fully explored. The goal of this study was to analyze whether depletion of aggregates from an AD brain suppresses its in vivo "seeding" capability. Removal of aggregates was performed by using the Aggregate Specific Reagent 1 (ASR1) compound which has been previously described to specifically bind misfolded species. Our results show that pre-treatment with ASR1-coupled magnetic beads reduces the in vivo misfolding inducing capability of an AD brain extract. These findings shed light respect to the active principle responsible for the prion-like spreading of Alzheimer's amyloid pathology and open the possibility of using seeds-capturing reagents as a promising target for AD treatment.
- Published
- 2014
- Full Text
- View/download PDF
17. Cross-seeding of misfolded proteins: implications for etiology and pathogenesis of protein misfolding diseases.
- Author
-
Rodrigo Morales, Ines Moreno-Gonzalez, and Claudio Soto
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Published
- 2013
- Full Text
- View/download PDF
18. Animal and Cellular Models of Alzheimer’s Disease: Progress, Promise, and Future Approaches
- Author
-
Laura Trujillo-Estrada, Juan Antonio García-León, Javier Vitorica, Antonia Gutierrez, David Baglietto-Vargas, Frank M. LaFerla, Raquel Sanchez-Varo, Cristina Nuñez-Diaz, Elisabeth Sanchez-Mejias, Jose Carlos Davila, and Ines Moreno-Gonzalez
- Subjects
0301 basic medicine ,Amyloid beta-Peptides ,business.industry ,General Neuroscience ,tau Proteins ,Neurodegenerative Diseases ,Mice, Transgenic ,Disease ,03 medical and health sciences ,Mice ,Disease Models, Animal ,030104 developmental biology ,0302 clinical medicine ,Human disease ,Alzheimer Disease ,Medicine ,Animals ,Humans ,Neurology (clinical) ,Disease progress ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Alzheimer’s disease (AD) is an incurable neurodegenerative disease affecting over 45 million people worldwide. Transgenic mouse models have made remarkable contributions toward clarifying the pathophysiological mechanisms behind the clinical manifestations of AD. However, the limited ability of these in vivo models to accurately replicate the biology of the human disease have precluded the translation of promising preclinical therapies to the clinic. In this review, we highlight several major pathogenic mechanisms of AD that were discovered using transgenic mouse models. Moreover, we discuss the shortcomings of current animal models and the need to develop reliable models for the sporadic form of the disease, which accounts for the majority of AD cases, as well as human cellular models to improve success in translating results into human treatments.
- Published
- 2022
19. Age-related immune alterations and cerebrovascular inflammation
- Author
-
Jose F. Moruno-Manchon, Louise D. McCullough, Carson Finger, Antonia Gutierrez, and Ines Moreno-Gonzalez
- Subjects
education.field_of_study ,business.industry ,Vascular disease ,Population ,Inflammation ,medicine.disease ,Systemic inflammation ,Pathophysiology ,Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,Immune system ,Immunology ,Medicine ,medicine.symptom ,business ,education ,Vascular dementia ,Molecular Biology ,Stroke - Abstract
Aging is associated with chronic systemic inflammation, which contributes to the development of many age-related diseases, including vascular disease. The world’s population is aging, leading to an increasing prevalence of both stroke and vascular dementia. The inflammatory response to ischemic stroke is critical to both stroke pathophysiology and recovery. Age is a predictor of poor outcomes after stroke. The immune response to stroke is altered in aged individuals, which contributes to the disparate outcomes between young and aged patients. In this review, we describe the current knowledge of the effects of aging on the immune system and the cerebral vasculature and how these changes alter the immune response to stroke and vascular dementia in animal and human studies. Potential implications of these age-related immune alterations on chronic inflammation in vascular disease outcome are highlighted.
- Published
- 2021
- Full Text
- View/download PDF
20. Requirement of brain interleukin33 for aquaporin4 expression in astrocytes and glymphatic drainage of abnormal tau
- Author
-
João Quevedo, Colin Carlock, Yahuan Lou, Jean Wu, Tatiana Barichello, William F Glass, April Ross, Junbo Shim, and Ines Moreno-Gonzalez
- Subjects
0301 basic medicine ,DNA repair ,Physiology ,Plaque, Amyloid ,tau Proteins ,medicine.disease_cause ,Article ,law.invention ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,law ,Alzheimer Disease ,medicine ,Animals ,Molecular Biology ,Aquaporin 4 ,Chemistry ,Autophagy ,Neurodegeneration ,Brain ,medicine.disease ,Interleukin-33 ,Peripheral ,Cell biology ,Psychiatry and Mental health ,030104 developmental biology ,Tauopathies ,Astrocytes ,Recombinant DNA ,Glymphatic system ,Tauopathy ,sense organs ,030217 neurology & neurosurgery ,Oxidative stress ,Neuroscience - Abstract
Defective aquaporin4 (AQP4)-mediated glymphatic drainage has been linked to tauopathy and amyloid plaque in Alzheimer’s disease. We now show that brain interleukin33 (IL33) is required for regulation of AQP4 expression in astrocytes, especially those at neuron-facing membrane domain (n-AQP4). First, IL33-deficient (Il33−/−) mice showed a loss of n-AQP4 after middle age, which coincided with a rapid accumulation of abnormal tau in neurons and a reduction in drainage of abnormal tau to peripheral tissues. Second, injection of recombinant IL33 induced robust expression of AQP4 at perivascular endfoot (p-AQP4) of astrocytes, but not n-AQP4, in Il33−/− brains. Although the increased p-AQP4 greatly accelerated drainage of intracerebroventricularly injected peptides, it did not substantially accelerate drainage of abnormal tau. These results suggest that p-AQP4 drives overall convective flow toward perivenous space, i.e., glymphatics, whereas n-AQP4 may generate an aqueous flow away from neurons to remove neuronal wastes, e.g., abnormal tau. We have previously shown the role of brain IL33 in DNA repair and autophagy in neurons with oxidative stress. Now, we show that IL33 deficiency also impairs glymphatic drainage. Defects in those mechanisms together may lead to chronic neurodegeneration and tauopathy at old age in IL33-deficient mice.
- Published
- 2021
21. Transgenic Mouse Models of Alzheimer's Disease: An Integrative Analysis
- Author
-
Raquel Sanchez-Varo, Marina Mejias-Ortega, Juan Jose Fernandez-Valenzuela, Cristina Nuñez-Diaz, Laura Caceres-Palomo, Laura Vegas-Gomez, Elisabeth Sanchez-Mejias, Laura Trujillo-Estrada, Juan Antonio Garcia-Leon, Ines Moreno-Gonzalez, Marisa Vizuete, Javier Vitorica, David Baglietto-Vargas, Antonia Gutierrez, Universidad de Sevilla. Departamento de Bioquímica y Biología Molecular, Instituto de Salud Carlos III, Junta de Andalucía, Ministerio de Ciencia e Innovación (MICIN). España, Alzheimer Association, Universidad de Málaga, European Commission, Ministerio de Ciencia e Innovación (España), and Alzheimer's Association
- Subjects
Amyloid ,Mice, Transgenic ,Plaque, Amyloid ,Catalysis ,Inorganic Chemistry ,Alzhéimer, Enfermedad de ,Amyloid beta-Protein Precursor ,Mice ,Alzheimer Disease ,Transgenic mice ,Animals ,Humans ,Physical and Theoretical Chemistry ,Neurodegeneration ,Molecular Biology ,Spectroscopy ,Aged ,Oligodendrocytes ,Organic Chemistry ,General Medicine ,Microglia Astrocytes ,Computer Science Applications ,Disease Models, Animal ,Astrocytes ,Microglia ,Tau ,Alzheimer’s disease - Abstract
Alzheimer's disease (AD) constitutes the most prominent form of dementia among elderly individuals worldwide. Disease modeling using murine transgenic mice was first initiated thanks to the discovery of heritable mutations in amyloid precursor protein (APP) and presenilins (PS) genes. However, due to the repeated failure of translational applications from animal models to human patients, along with the recent advances in genetic susceptibility and our current understanding on disease biology, these models have evolved over time in an attempt to better reproduce the complexity of this devastating disease and improve their applicability. In this review, we provide a comprehensive overview about the major pathological elements of human AD (plaques, tauopathy, synaptic damage, neuronal death, neuroinflammation and glial dysfunction), discussing the knowledge that available mouse models have provided about the mechanisms underlying human disease. Moreover, we highlight the pros and cons of current models, and the revolution offered by the concomitant use of transgenic mice and omics technologies that may lead to a more rapid improvement of the present modeling battery., This research was funded by INSTITUTO DE SALUD CARLOS III (ISCiii) of Spain, co-financed by FEDER funds from European Union, through grants PI21/00915 (to AG) and PI21/00914 (to JV); by JUNTA DE ANDALUCIA CONSEJERÍA DE ECONOMÍA Y CONOCIMIENTO through grants UMA18-FEDERJA-211 (to AG), UMA20-FEDERJA-104 (to IMG), P18-RT-2233 (to AG) and US-1262734 (to JV) co-financed by Programa Operativo FEDER 2014–2020 and CONSEJERIA DE SALUD grant PI-0276-2018 (to JAGL); by SPANISH MINISTER OF SCIENCE AND INNOVATION grant PID2019-108911RA-100 (to DBV), BEATRIZ GALINDO PROGRAM BAGAL18/00052 (to DBV), Alzheimer Association AARG-22-928219 (to DBV), grant PID2019-107090RA-100 (to IMG) and RAMON Y CAJAL PROGRAM RYC-2017-21879 (to IMG); and by MALAGA UNIVERSITY grant B1-2019_07 (to ESM), grant B1-2020_04 (to JAGL), grant B1-2019_06 (to IMG) and NASARD grant 27565 2018 (to IMG). M.M.-O. held a predoctoral contract from Malaga University, J.J.F.-V. held a postdoctoral contract from Malaga University, and E.S.-M. a postdoctoral contract (DOC_00251) from Junta de Andalucia.
- Published
- 2022
22. A near-infrared probe for detecting and interposing amyloid beta oligomerization in early Alzheimer's disease
- Author
-
Li Quan, Ines Moreno‐Gonzalez, Zhigang Xie, Nazaret Gamez, Laura Vegas‐Gomez, Qinyong Song, Jianhua Gu, Wenhai Lin, Ruben Gomez‐Gutierrez, and Tianfu Wu
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Abstract
The misfolding and deposition of amyloid beta (Aβ) in human brain is the main hallmark of Alzheimer's disease (AD) pathology. One of the drivers of Alzheimer´s pathogenesis is the production of soluble oligomeric Aβ, which could potentially serve as a biomarker of AD.Given that the diphenylalanine (FF) at the C-terminus of Aβ fragments plays a key role in inducing the AD pathology, based on the hydrophobic structure of FF, we synthesized a near-infrared BF2-dipyrrolmethane fluorescent imaging probe (NB) to detect both soluble and insoluble Aβ.We found that NB not only binds Aβ, particularly oligomeric Aβ, but also interposes self-assembly of Aβ through π-π interaction between NB and FF.This work holds great promise in the early detection of AD and may also provide an innovative approach to decelerate and even halt AD onset and progression.
- Published
- 2022
23. Use of neural precursors as a therapy to alleviate Alzheimer’s disease neuropathology and cognitive impairment
- Author
-
Nazaret Gamez Ruiz, Rodrigo Morales, Antonia Gutierrez, and Ines Moreno‐Gonzalez
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2021
- Full Text
- View/download PDF
24. G-quadruplexes Stabilization Upregulates CCN1 and Accelerates Aging in Cultured Cerebral Endothelial Cells
- Author
-
Brian Noh, Maria P. Blasco-Conesa, Yun-Ju Lai, Bhanu Priya Ganesh, Akihiko Urayama, Ines Moreno-Gonzalez, Sean P. Marrelli, Louise D. McCullough, and Jose Felix Moruno-Manchon
- Subjects
senescence ,G-quadruplex ,Geriatrics ,aging ,cardiovascular system ,RC952-954.6 ,CCN1 ,endothelial cells - Abstract
Senescence in the cerebral endothelium has been proposed as a mechanism that can drive dysfunction of the cerebral vasculature, which precedes vascular dementia. Cysteine-rich angiogenic inducer 61 (Cyr61/CCN1) is a matricellular protein secreted by cerebral endothelial cells (CEC). CCN1 induces senescence in fibroblasts. However, whether CCN1 contributes to senescence in CEC and how this is regulated requires further study. Aging has been associated with the formation of four-stranded Guanine-quadruplexes (G4s) in G-rich motifs of DNA and RNA. Stabilization of the G4 structures regulates transcription and translation either by upregulation or downregulation depending on the gene target. Previously, we showed that aged mice treated with a G4-stabilizing compound had enhanced senescence-associated (SA) phenotypes in their brains, and these mice exhibited enhanced cognitive deficits. A sequence in the 3′-UTR of the human CCN1 mRNA has the ability to fold into G4s in vitro. We hypothesize that G4 stabilization regulates CCN1 in cultured primary CEC and induces endothelial senescence. We used cerebral microvessel fractions and cultured primary CEC from young (4-months old, m/o) and aged (18-m/o) mice to determine CCN1 levels. SA phenotypes were determined by high-resolution fluorescence microscopy in cultured primary CEC, and we used Thioflavin T to recognize RNA-G4s for fluorescence spectra. We found that cultured CEC from aged mice exhibited enhanced levels of SA phenotypes, and higher levels of CCN1 and G4 stabilization. In cultured CEC, CCN1 induced SA phenotypes, such as SA β-galactosidase activity, and double-strand DNA damage. Furthermore, CCN1 levels were upregulated by a G4 ligand, and a G-rich motif in the 3′-UTR of the Ccn1 mRNA was folded into a G4. In conclusion, we demonstrate that CCN1 can induce senescence in cultured primary CEC, and we provide evidence that G4 stabilization is a novel mechanism regulating the SASP component CCN1.
- Published
- 2021
25. Longitudinal Assessment of Tau-Associated Pathology by 18F-THK5351 PET Imaging: A Histological, Biochemical, and Behavioral Study
- Author
-
Jonathan Schulz, Juan Jose Fernandez-Valenzuela, George A. Edwards, Claudio Soto, Antonia Gutierrez, Nazaret Gamez, Ines Moreno-Gonzalez, Paul E. Schulz, Omar Hasan, [Moreno-Gonzalez,I, Edwards III,GA, Hasan,O, Gamez,N, Schulz,JE, Soto,C, Schulz,PE] Department of Neurology, The University of Texas Health Science Center at Houston, Houston, USA. [Moreno-Gonzalez,I, Fernandez-Valenzuela,JJ, Gutierrez,A] Department of Cell Biology, Instituto de Investigacion Biomedica de Malaga-IBIMA, Faculty of Sicences, University of Malaga, Malaga, Spain. [Moreno-Gonzalez,I, Gutierrez,A] Networking Biomedical Research Center on Neurodegenerative Diseases (CIBERBED), Madrid, Spain., and This work was supported by a grant from the Alzheimer’s Association NIRG-394284, Department of Defense AZ160106, PID2019-107090RA-100, 27565 2018 NARSAD, and RYC-2017-21879 to I.M.G. PI18/01557 from Instituto de Salud Carlos III co-financed by FEDER funds EU, UMA18-FEDERJA-211, and P18-RT-2233 from Junta de Andalucia co-financed by FEDER 2014-2020 were granted to A.G. J.J.F.V. was supported by a PhD fellowship FPU program (Spanish Ministry of Science, Innovation and Universities).
- Subjects
Pathology ,Medicine (General) ,Diseases::Nervous System Diseases::Central Nervous System Diseases::Brain Diseases::Basal Ganglia Diseases::Supranuclear Palsy, Progressive [Medical Subject Headings] ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Diagnosis::Diagnostic Techniques and Procedures::Diagnostic Imaging::Tomography::Tomography, Emission-Computed::Positron-Emission Tomography [Medical Subject Headings] ,Clinical Biochemistry ,PET imaging ,Chemicals and Drugs::Inorganic Chemicals::Isotopes::Radioisotopes::Radioactive Tracers [Medical Subject Headings] ,Enfermedad de Alzheimer ,Corticobasal degeneration ,Diseases::Nervous System Diseases::Central Nervous System Diseases::Brain Diseases::Dementia::Alzheimer Disease [Medical Subject Headings] ,THK5351 ,Diseases::Nervous System Diseases::Central Nervous System Diseases::Brain Diseases::Dementia::Frontotemporal Lobar Degeneration::Frontotemporal Dementia [Medical Subject Headings] ,Tauopatías ,biology ,Anatomy::Cells::Cellular Structures::Intracellular Space::Cytoplasm::Cytoplasmic Structures::Cytoskeleton::Neurofibrils::Neurofibrillary Tangles [Medical Subject Headings] ,tracer ,Diseases::Nervous System Diseases::Central Nervous System Diseases::Brain Diseases::Dementia [Medical Subject Headings] ,Astrogliosis ,Tauopathy ,Cognitive impairment ,Histological assessment ,Técnicas histológicas ,biomarker ,Disfunción cognitiva ,Proteínas tau ,Alzheimer’s disease ,Frontotemporal dementia ,medicine.medical_specialty ,Tau protein ,Neuropathology ,Tomografía por emisión de positrones ,Article ,Progressive supranuclear palsy ,R5-920 ,Tracer ,Demencia ,mental disorders ,medicine ,Chemicals and Drugs::Biological Factors::Biological Markers [Medical Subject Headings] ,Dementia ,Chemicals and Drugs::Amino Acids, Peptides, and Proteins::Proteins::Cytoskeletal Proteins::Microtubule Proteins::Microtubule-Associated Proteins::tau Proteins [Medical Subject Headings] ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Diagnosis::Diagnostic Techniques and Procedures::Diagnostic Imaging::Image Interpretation, Computer-Assisted::Tomography, Emission-Computed::Positron-Emission Tomography [Medical Subject Headings] ,cognitive impairment ,Trazadores radiactivos ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Rodentia::Muridae::Murinae::Mice [Medical Subject Headings] ,business.industry ,histological assessment ,tauopathy ,Biomarker ,medicine.disease ,Diseases::Pathological Conditions, Signs and Symptoms::Pathologic Processes::Gliosis [Medical Subject Headings] ,Biomarcadores ,biology.protein ,Tau ,business ,dementia - Abstract
Several common and debilitating neurodegenerative disorders are characterized by the intracellular accumulation of neurofibrillary tangles (NFTs), which are composed of hyperphosphorylated tau protein. In Alzheimer’s disease (AD), NFTs are accompanied by extracellular amyloid-beta (Aβ), but primary tauopathy disorders are marked by the accumulation of tau protein alone, including forms of frontotemporal dementia (FTD), corticobasal degeneration (CBD), and progressive supranuclear palsy (PSP), among others. 18F-THK5351 has been reported to bind pathological tau as well as associated reactive astrogliosis. The goal of this study was to validate the ability of the PET tracer 18F-THK5351 to detect early changes in tau-related pathology and its relation to other pathological hallmarks. We demonstrated elevated in vivo 18F-THK5351 PET signaling over time in transgenic P301S tau mice from 8 months that had a positive correlation with histological and biochemical tau changes, as well as motor, memory, and learning impairment. This study indicates that 18F-THK5351 may help fill a critical need to develop PET imaging tracers that detect aberrant tau aggregation and related neuropathology in order to diagnose the onset of tauopathies, gain insights into their underlying pathophysiologies, and to have a reliable biomarker to follow during treatment trials.
- Published
- 2021
26. Aβ oligomers trigger necroptosis-mediated neurodegeneration via microglia activation in Alzheimer’s disease
- Author
-
Laura Vegas, Felipe A. Court, Ines Moreno-Gonzalez, Gabriel Quiroz, Claudio Soto, Javier Vitorica, Antonia Gutierrez, Marcela Escandón, Sebastian Jimenez, Nazaret Gamez Ruiz, and Natalia Salvadores
- Subjects
medicine.anatomical_structure ,Microglia ,Chemistry ,Necroptosis ,Neurodegeneration ,medicine ,Tumor necrosis factor alpha ,Alzheimer's disease ,medicine.disease ,Neuron death ,Beta (finance) ,Neuroscience ,Neuroinflammation - Abstract
Alzheimer’s disease (AD) is a major adult-onset neurodegenerative condition with no available treatment. Compelling reports point amyloid-β (Aβ) as the main etiologic agent that triggers AD. Although there is extensive evidence of detrimental crosstalk between Aβ and microglia that contributes to neuroinflammation in AD, the exact mechanism leading to neuron death remains unknown. Using postmortem human AD brain tissue, we show that Aβ pathology is associated with the necroptosis effector pMLKL. Moreover, we found that the burden of Aβo correlates with the expression of key markers of necroptosis activation. Additionally, inhibition of necroptosis by pharmacological or genetic means, reduce neurodegeneration and memory impairment triggered by Aβo in mice. Since microglial activation is emerging as a central driver for AD pathogenesis, we then tested the contribution of microglia to the mechanism of Aβo-mediated necroptosis activation in neurons. Using an in vitro model, we show that conditioned medium from Aβo-stimulated microglia elicited necroptosis in neurons through activation of TNF-α signaling, triggering extensive neurodegeneration. Notably, necroptosis inhibition provided significant neuronal protection. Together, these findings suggest that Aβo-mediated microglia stimulation in AD contributes to necroptosis activation in neurons and neurodegeneration. As necroptosis is a druggable degenerative mechanism, our findings might have important therapeutic implications to prevent the progression of AD.
- Published
- 2021
- Full Text
- View/download PDF
27. Age-related immune alterations and cerebrovascular inflammation
- Author
-
Carson E, Finger, Ines, Moreno-Gonzalez, Antonia, Gutierrez, Jose Felix, Moruno-Manchon, and Louise D, McCullough
- Subjects
Inflammation ,Stroke ,Aging ,Dementia, Vascular ,Animals ,Humans ,Aged - Abstract
Aging is associated with chronic systemic inflammation, which contributes to the development of many age-related diseases, including vascular disease. The world's population is aging, leading to an increasing prevalence of both stroke and vascular dementia. The inflammatory response to ischemic stroke is critical to both stroke pathophysiology and recovery. Age is a predictor of poor outcomes after stroke. The immune response to stroke is altered in aged individuals, which contributes to the disparate outcomes between young and aged patients. In this review, we describe the current knowledge of the effects of aging on the immune system and the cerebral vasculature and how these changes alter the immune response to stroke and vascular dementia in animal and human studies. Potential implications of these age-related immune alterations on chronic inflammation in vascular disease outcome are highlighted.
- Published
- 2021
28. Amyloid pathology arrangements in Alzheimer’s disease brains modulate in vivo seeding capability
- Author
-
Rodrigo Morales, Nelson Perez-Urrutia, Ines Moreno-Gonzalez, Claudia Duran-Aniotz, Nazaret Gamez, Laura Vegas-Gomez, Claudio Soto, [Duran-Aniotz,C, Moreno-Gonzalez,I, Gamez,N, Perez-Urrutia,N, Soto,C, Morales,R] Department of Neurology, The University of Texas Health Science Center at Houston, Fannin, St. Houston, USA. [Duran-Aniotz,C] Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile. [Duran-Aniotz,C] Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile. [Duran-Aniotz,C, Soto,C] Universidad de los Andes, Facultad de Medicina, Las Condes, Santiago, Chile. [Moreno-Gonzalez,I, Vegas-Gomez,L] Department of Cell Biology, Faculty of Sciences, University of Malaga-IBIMA, Malaga, Spain. [Moreno-Gonzalez,I] Networking Research Center On Neurodegenerative Diseases (CIBERNED), Madrid, Spain. [Moreno-Gonzalez,I, Morales,R] Centro Integrativo de Biologia Y Quimica Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago, Chile., and This work was supported by a grants from the NIH (R56AG061878 and RF1AG059321) to RM and CS, the Alzheimer’s Association (AARGD-18–566576 to RM and 2018-AARG-591107 to CDA), ANID/FONDEF ID20I10152 and ANID/FONDECYT 1210622 to CDA, and PID2019-107090RA-100, 27565 2018 NARSAD and RYC-2017–21879 to IMG.
- Subjects
Male ,Diseases::Nutritional and Metabolic Diseases::Metabolic Diseases::Proteostasis Deficiencies::Amyloidosis [Medical Subject Headings] ,Phenomena and Processes::Genetic Phenomena::Phenotype [Medical Subject Headings] ,Neurology ,Strains ,Disease ,lcsh:RC346-429 ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Primates::Haplorhini::Catarrhini::Hominidae::Humans [Medical Subject Headings] ,chemistry.chemical_compound ,Mice ,beta-Amiloides ,Organisms::Eukaryota::Animals [Medical Subject Headings] ,Pathology ,Organisms::Eukaryota::Animals::Animal Population Groups::Animals, Genetically Modified::Mice, Transgenic [Medical Subject Headings] ,Cognitive decline ,Persons::Persons::Age Groups::Adult::Aged [Medical Subject Headings] ,Aged, 80 and over ,Priones ,biology ,Amyloidosis ,Brain ,Patología ,Chemicals and Drugs::Amino Acids, Peptides, and Proteins::Proteins::Prions [Medical Subject Headings] ,Middle Aged ,Phenotype ,Thioflavin ,Female ,Amyloid-beta ,Alzheimer’s disease ,medicine.medical_specialty ,Diseases::Nervous System Diseases::Neurodegenerative Diseases::Tauopathies::Alzheimer Disease [Medical Subject Headings] ,Amyloid beta ,Prions ,Check Tags::Male [Medical Subject Headings] ,Mice, Transgenic ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,In vivo ,Alzheimer Disease ,medicine ,Animals ,Humans ,Diseases::Nervous System Diseases::Neurodegenerative Diseases::Prion Diseases [Medical Subject Headings] ,lcsh:Neurology. Diseases of the nervous system ,Aged ,Amyloid beta-Peptides ,Enfermedad de alzheimer ,Research ,Persons::Persons::Age Groups::Adult::Middle Aged [Medical Subject Headings] ,Chemicals and Drugs::Amino Acids, Peptides, and Proteins::Peptides::Amyloid beta-Peptides [Medical Subject Headings] ,Anatomy::Nervous System::Central Nervous System::Brain [Medical Subject Headings] ,medicine.disease ,Check Tags::Female [Medical Subject Headings] ,chemistry ,Persons::Persons::Age Groups::Adult::Aged::Aged, 80 and over [Medical Subject Headings] ,biology.protein ,Neurology (clinical) ,Neuroscience - Abstract
Amyloid-β (Aβ) misfolding is one of the hallmark pathological features of Alzheimer’s disease (AD). AD can manifest with diverse symptomatology including variable rates of cognitive decline, duration of clinical disease, and other detrimental changes. Several reports suggest that conformational diversity in misfolded Aβ is a leading factor for clinical variability in AD, analogous to what it has been described for prion strains in prion diseases. Notably, prion strains generate diverse patterns of misfolded protein deposition in the brains of affected individuals. Here, we tested the in vivo prion-like transmission features of four AD brains displaying particular patterns of amyloidosis. AD brains induced different phenotypes in recipient mice, as evaluated by their specific seeding activity, as well as the total amount of Aβ deposited surrounding vascular structures and the reactivity of amyloid pathology to thioflavin S. Our results support the notion that AD-subtypes are encoded in disease-associated Aβ. Further research exploring whether AD include a spectrum of different clinical conditions or syndromes may pave the way to personalized diagnosis and treatments. Supplementary Information The online version contains supplementary material available at 10.1186/s40478-021-01155-0.
- Published
- 2021
29. Transmission of cerebral amyloid pathology by peripheral administration of misfolded Aβ aggregates
- Author
-
Rodrigo, Morales, Javiera, Bravo-Alegria, Ines, Moreno-Gonzalez, Claudia, Duran-Aniotz, Nazaret, Gamez, George, Edwards Iii, and Claudio, Soto
- Subjects
Amyloid beta-Protein Precursor ,Disease Models, Animal ,Mice ,Amyloid beta-Peptides ,Alzheimer Disease ,Animals ,Brain ,Humans ,Mice, Transgenic ,Plaque, Amyloid ,Amyloidosis - Abstract
Previous reports showed that brain Aβ amyloidosis can be induced in animal models by exogenous administration of pre-formed aggregates. To date, only intra-peritoneal and intra-venous administrations are described as effective means to peripherally accelerate brain Aβ amyloidosis by seeding. Here, we show that cerebral accumulation of Aβ can be accelerated after exposing mouse models of Alzheimer's disease (AD) to Aβ seeds by different peripheral routes of administration, including intra-peritoneal and intra-muscular. Interestingly, animals receiving drops of brain homogenate laden with Aβ seeds in the eyes were efficiently induced. On the contrary, oral administration of large quantities of brain extracts from aged transgenic mice and AD patients did not have any effect in brain pathology. Importantly, pathological induction by peripheral administration of Aβ seeds generated a large proportion of aggregates in blood vessels, suggesting vascular transport. This information highlights the role of peripheral tissues and body fluids in AD-related pathological changes.
- Published
- 2020
30. Preventive and therapeutic reduction of amyloid deposition and behavioral impairments in a model of Alzheimer's disease by whole blood exchange
- Author
-
Akihiko Urayama, Ines Moreno-Gonzalez, Diego Morales-Scheihing, Vineetkumar Kharat, Sandra Pritzkow, and Claudio Soto
- Subjects
Amyloid beta-Peptides ,Brain ,Plaque, Amyloid ,Mice, Transgenic ,Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,Mice ,Protein Aggregates ,Disease Models, Animal ,Amyloid beta-Protein Precursor ,Alzheimer Disease ,Animals ,Humans ,Molecular Biology ,Aged - Abstract
Alzheimer's disease (AD) is the major form of dementia in the elderly population. The main neuropathological changes in AD patients are neuronal death, synaptic alterations, brain inflammation, and the presence of cerebral protein aggregates in the form of amyloid plaques and neurofibrillary tangles. Compelling evidence suggests that the misfolding, aggregation, and cerebral deposition of amyloid-beta (Aβ) plays a central role in the disease. Thus, prevention and removal of misfolded protein aggregates is considered a promising strategy to treat AD. In the present study, we describe that the development of cerebral amyloid plaques in a transgenic mice model of AD (Tg2576) was significantly reduced by 40-80% through exchanging whole blood with normal blood from wild type mice having the same genetic background. Importantly, such reduction resulted in improvement in spatial memory performance in aged Tg2576 mice. The exact mechanism by which blood exchange reduces amyloid pathology and improves memory is presently unknown, but measurements of Aβ in plasma soon after blood exchange suggest that mobilization of Aβ from the brain to blood may be implicated. Our results suggest that a target for AD therapy may exist in the peripheral circulation, which could open a novel disease-modifying intervention for AD.
- Published
- 2020
31. Infusion of neural precursors improves memory impairment in mouse models of Alzheimer’s disease
- Author
-
Ines Moreno-Gonzalez, Nazaret Gamez Ruiz, Claudio Soto, Ruben Gomez-Gutierrez, and Enrique Antonio Armijo Fuentes
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,business.industry ,Health Policy ,Medicine ,Memory impairment ,Neurology (clinical) ,Disease ,Geriatrics and Gerontology ,business ,Neuroscience - Published
- 2020
- Full Text
- View/download PDF
32. Detection of misfolded protein aggregated in HIV‐infected people
- Author
-
Benjamin B. Gelman, Olivia Calderon, Natalia Pessoa Rocha, Nazaret Gamez Ruiz, Ines Moreno-Gonzalez, and Claudio Soto
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,business.industry ,Health Policy ,Hiv infected ,Medicine ,Neurology (clinical) ,Neuropathology ,Geriatrics and Gerontology ,business ,Virology - Published
- 2020
- Full Text
- View/download PDF
33. Amyloid propagation in a sporadic model of Alzheimer's disease
- Author
-
Laura Trujillo-Estrada, Mohammad Shahnawaz, Antonia Gutierrez, Alessandra C. Martini, David Baglietto-Vargas, Marie Minh Thu Nguyen, Alwin Cheung, Ines Moreno-Gonzalez, Cristina Nuñez-Diaz, Stefania Forner, Celia da Cunha, Janine Pham Tran, Claudio Soto, Kelly Do Huynh, and Frank M. LaFerla
- Subjects
Pathology ,medicine.medical_specialty ,Amyloid ,Epidemiology ,business.industry ,Health Policy ,Disease ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,medicine ,Neurology (clinical) ,Geriatrics and Gerontology ,business - Published
- 2020
- Full Text
- View/download PDF
34. Enhancing microtubule stabilization rescues cognitive deficits and ameliorates pathological phenotype in an amyloidogenic Alzheimer’s disease model
- Author
-
Victoria Navarro, Javier Vitorica, Cristina Nuñez-Diaz, Sebastian Jimenez, Elisabeth Sanchez-Mejias, Angela Gomez-Arboledas, Clara Muñoz-Castro, Marisa Vizuete, Raquel Sanchez-Varo, Juan Jose Fernandez-Valenzuela, Ines Moreno-Gonzalez, Jose Carlos Davila, Antonia Gutierrez, Vanessa De Castro, Universidad de Sevilla. Departamento de Bioquímica y Biología Molecular, Instituto de Salud Carlos III (ISCiii) de España, European Commission (EC). Fondo Europeo de Desarrollo Regional (FEDER), Centro de Investigación Biomédica en Red (CIBERNED), Instituto de Salud Carlos III, European Commission, Centro Investigación Biomédica en Red Enfermedades Neurodegenerativas (España), Universidad de Málaga, Ministerio de Ciencia, Innovación y Universidades (España), [Fernandez-Valenzuela,JJ, Sanchez-Varo,R, De Castro,V, Sanchez-Mejias,E, Nuñez-Diaz,C, Gomez-Arboledas,A, Moreno-Gonzalez,I, Davila,JC, Gutierrez,A] Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga‑IBIMA, Facultad de Ciencias, Universidad de Málaga, Málaga, Spain. [Fernandez-Valenzuela,JJ, Muñoz-Castro,C, Navarro,V, Jimenez,S, Vizuete,M, Vitorica,J, Gutierrez,A] Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain. [Muñoz-Castro,C, Vitorica,J] Dpto. Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain. [Muñoz-Castro,C, Vitorica,J] Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocio/CSIC, Universidad de Sevilla, Sevilla, Spain., This study was supported by Instituto de Salud Carlos III (ISCiii) of Spain, co-financed by FEDER funds from European Union, through grants PI15/00796 and PI18/01557 (both to AG), PI15/00957 and PI18/01556 (both to JV), and CIBERNED (CB06/05/1116 to AG and CB06/05/0094 to JV), and by Malaga University grant PPIT.UMA.B1.2017/26 (to RSV). JJFV and CMC were supported by FPU PhD fellowships (Spanish Ministry of Science, Innovation and Universities). RSV held a postdoctoral contract from Malaga University. IMG is recipient of a senior postdoctoral contract from Ramon y Cajal Program (Spain Government).
- Subjects
Male ,Phenomena and Processes::Genetic Phenomena::Phenotype [Medical Subject Headings] ,Interneuronas ,Psychiatry and Psychology::Mental Disorders::Delirium, Dementia, Amnestic, Cognitive Disorders::Cognition Disorders [Medical Subject Headings] ,lcsh:Medicine ,Hippocampal formation ,Chemicals and Drugs::Organic Chemicals::Lactones::Macrolides::Epothilones [Medical Subject Headings] ,Axonal Transport ,Microtubules ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Primates::Haplorhini::Catarrhini::Hominidae::Humans [Medical Subject Headings] ,Mice ,Enfermedad de Alzheimer ,Organisms::Eukaryota::Animals [Medical Subject Headings] ,Organisms::Eukaryota::Animals::Animal Population Groups::Animals, Genetically Modified::Mice, Transgenic [Medical Subject Headings] ,lcsh:Science ,Anatomy::Nervous System::Neurons [Medical Subject Headings] ,Neurons ,Tauopatías ,Multidisciplinary ,Diseases::Nervous System Diseases::Neurodegenerative Diseases::Tauopathies [Medical Subject Headings] ,Neurodegeneration ,Brain ,Tubulin Modulators ,Phenotype ,Tauopathies ,Encéfalo ,Anatomy::Cells::Cellular Structures::Intracellular Space::Cytoplasm::Cytoplasmic Structures::Cytoskeleton::Microtubules [Medical Subject Headings] ,GABAergic ,Female ,Intracellular ,Amyloid ,Diseases::Nervous System Diseases::Neurodegenerative Diseases::Tauopathies::Alzheimer Disease [Medical Subject Headings] ,Transporte axonal ,Chemicals and Drugs::Chemical Actions and Uses::Pharmacologic Actions::Molecular Mechanisms of Pharmacological Action::Mitosis Modulators::Antimitotic Agents::Tubulin Modulators [Medical Subject Headings] ,Check Tags::Male [Medical Subject Headings] ,Mice, Transgenic ,Biology ,Neuroprotection ,Article ,Interneurons ,Alzheimer Disease ,Microtubule ,Extracellular ,medicine ,Animals ,Humans ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Rodentia::Muridae::Murinae::Mice [Medical Subject Headings] ,Phenomena and Processes::Cell Physiological Phenomena::Cell Physiological Processes::Cytoplasmic Streaming::Axonal Transport [Medical Subject Headings] ,Amiloide ,lcsh:R ,medicine.disease ,Cellular neuroscience ,Diseases::Animal Diseases::Disease Models, Animal [Medical Subject Headings] ,Disease Models, Animal ,Check Tags::Female [Medical Subject Headings] ,Epothilones ,Axoplasmic transport ,Diseases of the nervous system ,lcsh:Q ,Cognition Disorders ,Neuroscience - Abstract
In Alzheimer’s disease (AD), and other tauopathies, microtubule destabilization compromises axonal and synaptic integrity contributing to neurodegeneration. These diseases are characterized by the intracellular accumulation of hyperphosphorylated tau leading to neurofibrillary pathology. AD brains also accumulate amyloid-beta (Aβ) deposits. However, the effect of microtubule stabilizing agents on Aβ pathology has not been assessed so far. Here we have evaluated the impact of the brain-penetrant microtubule-stabilizing agent Epothilone D (EpoD) in an amyloidogenic model of AD. Three-month-old APP/PS1 mice, before the pathology onset, were weekly injected with EpoD for 3 months. Treated mice showed significant decrease in the phospho-tau levels and, more interesting, in the intracellular and extracellular hippocampal Aβ accumulation, including the soluble oligomeric forms. Moreover, a significant cognitive improvement and amelioration of the synaptic and neuritic pathology was found. Remarkably, EpoD exerted a neuroprotective effect on SOM-interneurons, a highly AD-vulnerable GABAergic subpopulation. Therefore, our results suggested that EpoD improved microtubule dynamics and axonal transport in an AD-like context, reducing tau and Aβ levels and promoting neuronal and cognitive protection. These results underline the existence of a crosstalk between cytoskeleton pathology and the two major AD protein lesions. Therefore, microtubule stabilizers could be considered therapeutic agents to slow the progression of both tau and Aβ pathology., This study was supported by Instituto de Salud Carlos III (ISCiii) of Spain, co-financed by FEDER funds from European Union, through grants PI15/00796 and PI18/01557 (both to AG), PI15/00957 and PI18/01556 (both to JV), and CIBERNED (CB06/05/1116 to AG and CB06/05/0094 to JV); by Malaga University grant PPIT.UMA.B1.2017/26 (to RSV). JJFV and CMC were supported by FPU PhD fellowships (Spanish Ministry of Science, Innovation and Universities). RSV held a postdoctoral contract from Malaga University. IMG is recipient of a senior postdoctoral contract from Ramon y Cajal Program (Spain Government).
- Published
- 2020
- Full Text
- View/download PDF
35. Distinct disease‐sensitive GABAergic neurons in the perirhinal cortex of Alzheimer's mice and patients
- Author
-
Jose Carlos Davila, Javier Vitorica, Juan Antonio García-León, Laura Trujillo-Estrada, Marina Mejias-Ortega, Angela Gomez-Arboledas, Raquel Sanchez-Varo, Elisabeth Sanchez-Mejias, Juan Jose Fernandez-Valenzuela, Antonia Gutierrez, Ines Moreno-Gonzalez, Cristina Nuñez-Diaz, David Baglietto-Vargas, Instituto de Salud Carlos III, European Commission, Centro Investigación Biomédica en Red Enfermedades Neurodegenerativas (España), Junta de Andalucía, Universidad de Málaga, Ministerio de Ciencia, Innovación y Universidades (España), Universidad de Sevilla. Departamento de Bioquímica y Biología Molecular, European Commission (EC). Fondo Europeo de Desarrollo Regional (FEDER), and Centro de Investigaciones Biomédicas en Red (CIBERNED)
- Subjects
0301 basic medicine ,transentorhinal cortex ,Mice ,GABA ,0302 clinical medicine ,parvalbumin ,Perirhinal cortex ,GABAergic Neurons ,Research Articles ,Parvalbumin ,Aged, 80 and over ,education.field_of_study ,biology ,General Neuroscience ,Neurodegeneration ,Human brain ,Middle Aged ,medicine.anatomical_structure ,GABAergic ,Somatostatin ,Research Article ,Interneuron ,Population ,Mice, Transgenic ,interneuron ,somatostatin ,Pathology and Forensic Medicine ,03 medical and health sciences ,Alzheimer Disease ,Interneurons ,Transgenic mouse ,medicine ,Dementia ,Animals ,Humans ,Transentorhinal cortex ,education ,human brain ,Aged ,Perirhinal Cortex ,medicine.disease ,transgenic mouse ,Disease Models, Animal ,030104 developmental biology ,biology.protein ,Alzheimer ,Neurology (clinical) ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Neuronal loss is the best neuropathological substrate that correlates with cortical atrophy and dementia in Alzheimer's disease (AD). Defective GABAergic neuronal functions may lead to cortical network hyperactivity and aberrant neuronal oscillations and in consequence, generate a detrimental alteration in memory processes. In this study, using immunohistochemical and stereological approaches, we report that the two major and non‐overlapping groups of inhibitory interneurons (SOM‐cells and PV‐cells) displayed distinct vulnerability in the perirhinal cortex of APP/PS1 mice and AD patients. SOM‐positive neurons were notably sensitive and exhibited a dramatic decrease in the perirhinal cortex of 6‐month‐old transgenic mice (57% and 61% in areas 36 and 35, respectively) and, most importantly, in AD patients (91% in Braak V–VI cases). In addition, this interneuron degenerative process seems to occur in parallel, and closely related, with the progression of the amyloid pathology. However, the population expressing PV was unaffected in APP/PS1 mice while in AD brains suffered a pronounced and significant loss (69%). As a key component of cortico‐hippocampal networks, the perirhinal cortex plays an important role in memory processes, especially in familiarity‐based memory recognition. Therefore, disrupted functional connectivity of this cortical region, as a result of the early SOM and PV neurodegeneration, might contribute to the altered brain rhythms and cognitive failures observed in the initial clinical phase of AD patients. Finally, these findings highlight the failure of amyloidogenic AD models to fully recapitulate the selective neuronal degeneration occurring in humans., This study was supported by Instituto de Salud Carlos III (ISCiii) of Spain, co‐financed by FEDER funds from European Union, through grants PI18/01557 (to AG) and PI18/01556 (to JV), and CIBERNED (to AG and JV), by Consejería de Economía, Innovación, Ciencia y Empleo, Junta de Andalucia Proyecto de Excelencia (CTS‐2035) (to JV and AG); by Malaga University grant PPIT.UMA.B1.2017/26 (to RSV). CND and JJFV were supported by FPI (Junta Andalucía) and FPU (Spanish Ministry of Science, Innovation and Universities) PhD fellowships, respectively. MMO held a Garantia Juvenil (Junta Andalucia) contract. RSV and JAGL held a postdoctoral contract from the University of Malaga, and AGA from CIBERNED.
- Published
- 2020
36. Inhibition of protein misfolding and aggregation by natural phenolic compounds
- Author
-
Ines Moreno-Gonzalez, Karina Cuanalo-Contreras, Charles E. Mays, Claudio Soto, El Akrem Hayouni, and Zohra Dhouafli
- Subjects
0301 basic medicine ,Protein Folding ,Parkinson's disease ,Amyloid beta ,Protein Misfolding Disorder ,Amylin ,Disease ,Protein Aggregation, Pathological ,Prion Diseases ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,Phenols ,medicine ,Animals ,Humans ,Proteostasis Deficiencies ,Amyotrophic lateral sclerosis ,Molecular Biology ,Pharmacology ,Alpha-synuclein ,Biological Products ,biology ,Amyloidosis ,Cell Biology ,medicine.disease ,030104 developmental biology ,Diabetes Mellitus, Type 2 ,chemistry ,Biochemistry ,biology.protein ,Molecular Medicine ,Protein folding ,030217 neurology & neurosurgery - Abstract
Protein misfolding and aggregation into fibrillar deposits is a common feature of a large group of degenerative diseases affecting the central nervous system or peripheral organs, termed protein misfolding disorders (PMDs). Despite their established toxic nature, clinical trials aiming to reduce misfolded aggregates have been unsuccessful in treating or curing PMDs. An interesting possibility for disease intervention is the regular intake of natural food or herbal extracts, which contain active molecules that inhibit aggregation or induce the disassembly of misfolded aggregates. Among natural compounds, phenolic molecules are of particular interest, since most have dual activity as amyloid aggregation inhibitors and antioxidants. In this article, we review many phenolic natural compounds which have been reported in diverse model systems to have the potential to delay or prevent the development of various PMDs, including Alzheimer's and Parkinson's diseases, prion diseases, amyotrophic lateral sclerosis, systemic amyloidosis, and type 2 diabetes. The lower toxicity of natural compounds compared to synthetic chemical molecules suggest that they could serve as a good starting point to discover protein misfolding inhibitors that might be useful for the treatment of various incurable diseases.
- Published
- 2018
- Full Text
- View/download PDF
37. Treatment with a non-toxic, self-replicating anti-prion delays or prevents prion disease in vivo
- Author
-
Fabio Moda, Rodrigo Diaz-Espinoza, Luis Concha-Marambio, Claudio Soto, Ines Moreno-Gonzalez, and Rodrigo Morales
- Subjects
0301 basic medicine ,Gene isoform ,Prions ,animal diseases ,Brain damage ,Disease ,Protein Engineering ,Prion Diseases ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,In vivo ,medicine ,Animals ,Proteostasis Deficiencies ,Molecular Biology ,Infectivity ,Mesocricetus ,biology ,Brain ,Protein engineering ,biology.organism_classification ,Virology ,nervous system diseases ,3. Good health ,Psychiatry and Mental health ,030104 developmental biology ,Toxicity ,medicine.symptom - Abstract
Transmissible spongiform encephalopathies (TSEs) are fatal neurological disorders caused by prions, which are composed of a misfolded protein (PrPSc) that self-propagates in the brain of infected individuals by converting the normal prion protein (PrPC) into the pathological isoform. Here, we report a novel experimental strategy for preventing prion disease based on producing a self-replicating, but innocuous PrPSc-like form, termed anti-prion, which can compete with the replication of pathogenic prions. Our results show that a prophylactic inoculation of prion-infected animals with an anti-prion delays the onset of the disease and in some animals completely prevents the development of clinical symptoms and brain damage. The data indicate that a single injection of the anti-prion eliminated ~99% of the infectivity associated to pathogenic prions. Furthermore, this treatment caused significant changes in the profile of regional PrPSc deposition in the brains of animals that were treated, but still succumbed to the disease. Our findings provide new insights for a mechanistic understanding of prion replication and support the concept that prion replication can be separated from toxicity, providing a novel target for therapeutic intervention.
- Published
- 2017
- Full Text
- View/download PDF
38. Amyloid-beta and tau pathology following repetitive mild traumatic brain injury
- Author
-
George A. Edwards, Claudio Soto, and Ines Moreno-Gonzalez
- Subjects
0301 basic medicine ,Amyloid beta ,Traumatic brain injury ,Biophysics ,tau Proteins ,Disease ,Protein aggregation ,Biochemistry ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Brain Injuries, Traumatic ,Animals ,Humans ,Medicine ,Molecular Biology ,Pathological ,Neuroinflammation ,Amyloid beta-Peptides ,biology ,business.industry ,Neurodegenerative Diseases ,Cell Biology ,medicine.disease ,Chronic traumatic encephalopathy ,030104 developmental biology ,biology.protein ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Neurodegenerative diseases are characterized by distinctive neuropathological alterations, including the cerebral accumulation of misfolded protein aggregates, neuroinflammation, synaptic dysfunction, and neuronal loss, along with behavioral impairments. Traumatic brain injury (TBI) is believed to be an important risk factor for certain neurodegenerative diseases, such as Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). TBI represents a ubiquitous problem in the world and could play a major role in the pathogenesis and etiology of AD or CTE later in life. TBI events appear to trigger and exacerbate some of the pathological processes in these diseases, in particular, the formation and accumulation of misfolded protein aggregates composed of amyloid-beta (Aβ) and tau. Here, we describe the relationship between repetitive mild TBI and the development of Aβ and tau pathology in patients affected by AD or CTE on the basis of epidemiological and pathological studies in human cases, and a thorough overview of data obtained in experimental animal models. We also discuss the possibility that TBI may contribute to initiate the formation of misfolded oligomeric species that may subsequently spread the pathology through a prion-like process of seeding of protein misfolding.
- Published
- 2017
- Full Text
- View/download PDF
39. Risk Factors for Alzheimer’s Disease
- Author
-
David Baglietto-Vargas, Rodrigo Morales, Ines Moreno-Gonzalez, and Raquel Sanchez-Varo
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,Genetic variation ,medicine ,Disease ,business - Published
- 2020
- Full Text
- View/download PDF
40. Peripheral Delivery of Neural Precursor Cells Ameliorates Parkinson’s Disease-Associated Pathology
- Author
-
Nazaret Gamez, Claudio Soto, Paul E. Schulz, Carlos Kramm, Ines Moreno-Gonzalez, Enrique Armijo, George A. Edwards, Kathleen Taylor-Presse, and Rodrigo Morales
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Parkinson's disease ,medicine.medical_treatment ,neuronal precursors ,Neurotoxins ,Neuropathology ,medicine.disease_cause ,Article ,Mice ,Neural Stem Cells ,stem cells ,Precursor cell ,medicine ,Animals ,lcsh:QH301-705.5 ,Embryonic Stem Cells ,clinical symptoms ,therapy ,business.industry ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Parkinson Disease ,General Medicine ,Stem-cell therapy ,medicine.disease ,Embryonic stem cell ,nervous system diseases ,Mice, Inbred C57BL ,Disease Models, Animal ,Oxidative Stress ,lcsh:Biology (General) ,inflammation ,1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine ,intravenous ,Nerve Degeneration ,Parkinson’s disease ,Female ,Stem cell ,business ,Oxidative stress ,Stem Cell Transplantation - Abstract
Parkinson&rsquo, s disease (PD) is a progressive neurodegenerative disorder characterized by loss of motor control due to a wide loss of dopaminergic neurons along the nigro-striatal pathway. Some of the mechanisms that contribute to this cell death are inflammation, oxidative stress, and misfolded alpha-synuclein-induced toxicity. Current treatments are effective at managing the early motor symptoms of the disease, but they become ineffective over time and lead to adverse effects. Previous research using intracerebral stem cell therapy for treatment of PD has provided promising results, however, this method is very invasive and is often associated with unacceptable side effects. In this study, we used an MPTP-injected mouse model of PD and intravenously administered neural precursors (NPs) obtained from mouse embryonic and mesenchymal stem cells. Clinical signs and neuropathology were assessed. Female mice treated with NPs had improved motor function and reduction in the neuroinflammatory response. In terms of safety, there were no tumorigenic formations or any detectable adverse effect after treatment. Our results suggest that peripheral administration of stem cell-derived NPs may be a promising and safe therapy for the recovery of impaired motor function and amelioration of brain pathology in PD.
- Published
- 2019
41. Natural Products as Modulators of the Proteostasis Machinery: Implications in Neurodegenerative Diseases
- Author
-
Ines Moreno-Gonzalez and Karina Cuanalo-Contreras
- Subjects
autophagy ,Review ,Disease ,Protein Homeostasis ,Biology ,Neuroprotection ,Catalysis ,Inorganic Chemistry ,lcsh:Chemistry ,Alzheimer Disease ,medicine ,natural compounds ,Animals ,Humans ,chaperones ,Physical and Theoretical Chemistry ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,Biological Products ,proteostasis ,Organic Chemistry ,Autophagy ,Neurodegeneration ,neurodegeneration ,Parkinson Disease ,General Medicine ,unfolded protein response ,medicine.disease ,Computer Science Applications ,Proteostasis ,Neuroprotective Agents ,lcsh:Biology (General) ,lcsh:QD1-999 ,Unfolded protein response ,Neuroscience ,Function (biology) ,ubiquitin-proteasome - Abstract
Proteins play crucial and diverse roles within the cell. To exert their biological function they must fold to acquire an appropriate three-dimensional conformation. Once their function is fulfilled, they need to be properly degraded to hamper any possible damage. Protein homeostasis or proteostasis comprises a complex interconnected network that regulates different steps of the protein quality control, from synthesis and folding, to degradation. Due to the primary role of proteins in cellular function, the integrity of this network is critical to assure functionality and health across lifespan. Proteostasis failure has been reported in the context of aging and neurodegeneration, such as Alzheimer’s and Parkinson’s disease. Therefore, targeting the proteostasis elements emerges as a promising neuroprotective therapeutic approach to prevent or ameliorate the progression of these disorders. A variety of natural products are known to be neuroprotective by protein homeostasis interaction. In this review, we will focus on the current knowledge regarding the use of natural products as modulators of different components of the proteostasis machinery within the framework of age-associated neurodegenerative diseases.
- Published
- 2019
42. O5‐04‐02: TRAUMATIC BRAIN INJURY INDUCES TAU AGGREGATION AND SPREADING
- Author
-
Pramod K. Dash, Ines Moreno-Gonzalez, Claudio Soto, Jing Zhao, and George A. Edwards
- Subjects
0303 health sciences ,Epidemiology ,Traumatic brain injury ,business.industry ,Health Policy ,medicine.disease ,03 medical and health sciences ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Developmental Neuroscience ,medicine ,Neurology (clinical) ,Geriatrics and Gerontology ,business ,Neuroscience ,030217 neurology & neurosurgery ,030304 developmental biology - Published
- 2019
- Full Text
- View/download PDF
43. The necroptosis machinery mediates axonal degeneration in a model of Parkinson disease
- Author
-
Paulina Soto, Nazaret Gamez, Felipe A. Court, Alexis Martinez, Claudio Soto, Natalia Salvadores, Maritza Oñate, Cristian Saquel, Alejandra Catenaccio, Claudio Hetz, and Ines Moreno-Gonzalez
- Subjects
0301 basic medicine ,Programmed cell death ,Necroptosis ,Substantia nigra ,Striatum ,Degeneration (medical) ,Motor Activity ,Biology ,Article ,chemistry.chemical_compound ,03 medical and health sciences ,RIPK1 ,0302 clinical medicine ,Dopamine ,Neurites ,medicine ,Animals ,Humans ,Phosphorylation ,Oxidopamine ,Molecular Biology ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Dopaminergic Neurons ,Dopaminergic ,Correction ,Parkinson Disease ,Cell Biology ,Axons ,Mice, Inbred C57BL ,Substantia Nigra ,Disease Models, Animal ,030104 developmental biology ,chemistry ,nervous system ,030220 oncology & carcinogenesis ,Receptor-Interacting Protein Serine-Threonine Kinases ,Nerve Degeneration ,Protein Kinases ,Neuroscience ,Biomarkers ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative condition, characterized by motor impairment due to the progressive degeneration of dopaminergic neurons in the substantia nigra and depletion of dopamine release in the striatum. Accumulating evidence suggest that degeneration of axons is an early event in the disease, involving destruction programs that are independent of the survival of the cell soma. Necroptosis, a programmed cell death process, is emerging as a mediator of neuronal loss in models of neurodegenerative diseases. Here, we demonstrate activation of necroptosis in postmortem brain tissue from PD patients and in a toxin-based mouse model of the disease. Inhibition of key components of the necroptotic pathway resulted in a significant delay of 6-hydroxydopamine dependent axonal degeneration of dopaminergic and cortical neurons in vitro. Genetic ablation of necroptosis mediators MLKL and RIPK3, as well as pharmacological inhibition of RIPK1 in vivo, decreased dopaminergic neuron degeneration, improving motor performance. Together, these findings suggest that axonal degeneration in PD is mediated by the necroptosis machinery, a process here referred to as necroaxoptosis, a druggable pathway to target dopaminergic neuronal loss.
- Published
- 2019
- Full Text
- View/download PDF
44. Induction of IAPP amyloid deposition and associated diabetic abnormalities by a prion-like mechanism
- Author
-
Diego Morales-Scheihing, Mohammad Shahnawaz, A. Osama Gaber, Claudio Soto, Cesar Gonzalez, Ines Moreno-Gonzalez, Abhisek Mukherjee, Daniel W. Fraga, Nicolas Mendez, Natalia Salvadores, Omaima M. Sabek, and Kathleen Taylor-Presse
- Subjects
0301 basic medicine ,Genetically modified mouse ,Male ,medicine.medical_specialty ,endocrine system ,endocrine system diseases ,Amyloid ,Prions ,Immunology ,Endogeny ,Mice, Transgenic ,Protein aggregation ,Article ,03 medical and health sciences ,Islets of Langerhans ,Mice ,Protein Aggregates ,0302 clinical medicine ,In vivo ,Internal medicine ,medicine ,Immunology and Allergy ,Animals ,Humans ,Proteostasis Deficiencies ,Research Articles ,geography ,geography.geographical_feature_category ,biology ,Chemistry ,Islet ,In vitro ,Islet Amyloid Polypeptide ,030104 developmental biology ,Endocrinology ,Diabetes Mellitus, Type 2 ,biology.protein ,Female ,Antibody ,030217 neurology & neurosurgery - Abstract
In this article, Mukherjee et al. show that the pathologic and clinical alterations of type 2 diabetes can be induced in vitro and in vivo by prion-like transmission of IAPP misfolded aggregates, supporting an important role for IAPP aggregation in the disease., Although a large proportion of patients with type 2 diabetes (T2D) accumulate misfolded aggregates composed of the islet amyloid polypeptide (IAPP), its role in the disease is unknown. Here, we show that pancreatic IAPP aggregates can promote the misfolding and aggregation of endogenous IAPP in islet cultures obtained from transgenic mouse or healthy human pancreas. Islet homogenates immunodepleted with anti-IAPP–specific antibodies were not able to induce IAPP aggregation. Importantly, intraperitoneal inoculation of pancreatic homogenates containing IAPP aggregates into transgenic mice expressing human IAPP dramatically accelerates IAPP amyloid deposition, which was accompanied by clinical abnormalities typical of T2D, including hyperglycemia, impaired glucose tolerance, and a substantial reduction on β cell number and mass. Finally, induction of IAPP deposition and diabetic abnormalities were also induced in vivo by administration of IAPP aggregates prepared in vitro using pure, synthetic IAPP. Our findings suggest that some of the pathologic and clinical alterations of T2D might be transmissible through a similar mechanism by which prions propagate in prion diseases.
- Published
- 2017
45. Molecular interaction between type 2 diabetes and Alzheimer’s disease through cross-seeding of protein misfolding
- Author
-
Ines Moreno-Gonzalez, Claudio Soto, Mohammad Shahnawaz, Rodrigo Diaz-Espinoza, George A. Edwards, and Natalia Salvadores
- Subjects
0301 basic medicine ,Genetically modified mouse ,medicine.medical_specialty ,Protein Folding ,endocrine system ,Amyloid ,Amyloid beta ,Transgene ,Amylin ,Mice, Transgenic ,Plaque, Amyloid ,tau Proteins ,Protein aggregation ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,Risk Factors ,Alzheimer Disease ,Internal medicine ,medicine ,Animals ,Humans ,Molecular Biology ,Amyloid beta-Peptides ,biology ,Pancreatic islets ,Brain ,medicine.disease ,3. Good health ,Cell biology ,Islet Amyloid Polypeptide ,Psychiatry and Mental health ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Diabetes Mellitus, Type 2 ,biology.protein ,Alzheimer's disease ,030217 neurology & neurosurgery - Abstract
Numerous epidemiological studies have shown a significantly higher risk for development of Alzheimer's disease (AD) in patients affected by type 2 diabetes (T2D), but the molecular mechanism responsible for this association is presently unknown. Both diseases are considered protein misfolding disorders associated with the accumulation of protein aggregates; amyloid-beta (Aβ) and tau in the brain during AD, and islet amyloid polypeptide (IAPP) in pancreatic islets in T2D. Formation and accumulation of these proteins follows a seeding-nucleation model, where a misfolded aggregate or 'seed' promotes the rapid misfolding and aggregation of the native protein. Our underlying hypothesis is that misfolded IAPP produced in T2D potentiates AD pathology by cross-seeding Aβ, providing a molecular explanation for the link between these diseases. Here, we examined how misfolded IAPP affects Aβ aggregation and AD pathology in vitro and in vivo. We observed that addition of IAPP seeds accelerates Aβ aggregation in vitro in a seeding-like manner and the resulting fibrils are composed of both peptides. Transgenic animals expressing both human proteins exhibited exacerbated AD-like pathology compared with AD transgenic mice or AD transgenic animals with type 1 diabetes (T1D). Remarkably, IAPP colocalized with amyloid plaques in brain parenchymal deposits, suggesting that these peptides may directly interact and aggravate the disease. Furthermore, inoculation of pancreatic IAPP aggregates into the brains of AD transgenic mice resulted in more severe AD pathology and significantly greater memory impairments than untreated animals. These data provide a proof-of-concept for a new disease mechanism involving the interaction of misfolded proteins through cross-seeding events which may contribute to accelerate or exacerbate disease pathogenesis. Our findings could shed light on understanding the linkage between T2D and AD, two of the most prevalent protein misfolding disorders.
- Published
- 2017
46. Impaired Peripheral Lymphatic Function and Cerebrospinal Fluid Outflow in a Mouse Model of Alzheimer’s Disease
- Author
-
Olivia Calderon, George A. Edwards, Eva M. Sevick-Muraca, Banghe Zhu, Sunkuk Kwon, Ines Moreno-Gonzalez, Nazaret Gamez, Kathleen Taylor-Presse, Claudio Soto, and Fred Christian Velasquez
- Subjects
0301 basic medicine ,Genetically modified mouse ,Male ,Pathology ,medicine.medical_specialty ,Mice, Transgenic ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Cerebrospinal fluid ,Alzheimer Disease ,medicine ,Animals ,Humans ,Injections, Spinal ,Cerebrospinal Fluid ,Lymphatic Vessels ,Brain Chemistry ,Spectroscopy, Near-Infrared ,business.industry ,General Neuroscience ,Brain ,General Medicine ,Peripheral ,Psychiatry and Mental health ,Clinical Psychology ,030104 developmental biology ,Lymphatic system ,chemistry ,Outflow ,Female ,Geriatrics and Gerontology ,business ,Peripheral lymph ,Indocyanine green ,Lymphangion ,030217 neurology & neurosurgery ,Evans Blue - Abstract
Cerebrospinal fluid (CSF) outflow from the brain occurs through absorption into the arachnoid villi and, more predominantly, through meningeal and olfactory lymphatics that ultimately drain into the peripheral lymphatics. Impaired CSF outflow has been postulated as a contributing mechanism in Alzheimer's disease (AD). Herein we conducted near-infrared fluorescence imaging of CSF outflow into the peripheral lymph nodes (LNs) and of peripheral lymphatic function in a transgenic mouse model of AD (5XFAD) and wild-type (WT) littermates. CSF outflow was assessed from change in fluorescence intensity in the submandibular LNs as a function of time following bolus, an intrathecal injection of indocyanine green (ICG). Peripheral lymphatic function was measured by assessing lymphangion contractile function in lymphatics draining into the popliteal LN following intradermal ICG injection in the dorsal aspect of the hind paw. The results show 1) significantly impaired CSF outflow into the submandibular LNs of 5XFAD mice and 2) reduced contractile frequency in the peripheral lymphatics as compared to WT mice. Impaired CSF clearance was also evidenced by reduction of fluorescence on ventral surfaces of extracted brains of 5XFAD mice at euthanasia. These results support the hypothesis that lymphatic congestion caused by reduced peripheral lymphatic function could limit CSF outflow and may contribute to the cause and/or progression of AD.
- Published
- 2019
47. Monitoring the Formation of Amyloid Oligomers Using Photoluminescence Anisotropy
- Author
-
Fernando Godoy, Angel A. Martí, Bo Jiang, Erick I. Saavedra Flores, Amir Aliyan, Ines Moreno-Gonzalez, Rodrigo R. Maldonado, Ghibom Bhak, Javier Montenegro, Ashleigh D. Smith McWilliams, Nathan P. Cook, Mohammad Shahnawaz, Nicolas Mendez, Andrea Augustine, Universidade de Santiago de Compostela. Centro de Investigación en Química Biolóxica e Materiais Moleculares, and Universidade de Santiago de Compostela. Departamento de Química Orgánica
- Subjects
Amyloid ,Photoluminescence ,Polymers ,chemistry.chemical_element ,010402 general chemistry ,Photochemistry ,01 natural sciences ,Biochemistry ,Catalysis ,Colloid and Surface Chemistry ,Molecule ,Anisotropy ,Rotational correlation time ,Chemistry ,Aβ oligomers ,General Chemistry ,Photochemical Processes ,0104 chemical sciences ,3. Good health ,Ruthenium ,Luminescent Measurements ,Ruthenium Compounds ,Steady state (chemistry) - Abstract
The formation of oligomeric soluble aggregates is related to the toxicity of amyloid peptides and proteins. In this manuscript, we report the use of a ruthenium polypyridyl complex ([Ru(bpy)2(dpqp)]2+) to track the formation of amyloid oligomers at different times using photoluminescence anisotropy. This technique is sensitive to the rotational correlation time of the molecule under study, which is consequently related to the size of the molecule. [Ru(bpy)2(dpqp)]2+ presents anisotropy values of zero when free in solution (due to its rapid rotation and long lifetime) but larger values as the size and concentration of amyloid-β (Aβ) oligomers increase. Our assays show that Aβ forms oligomers immediately after the assay is started, reaching a steady state at ∼48 h. SDS–PAGE, DLS, and TEM were used to confirm and characterize the formation of oligomers. Our experiments show that the rate of formation for Aβ oligomers is temperature dependent, with faster rates as the temperature of the assay is increased. The probe was also effective in monitoring the formation of α-synuclein oligomers at different times AAM thanks the Welch Foundation (Grant C-1743) and JM thanks AEI (SAF2017-89890-R), ERC (DYNAP-677786) and HFSP (RGY0066/2017) for financial support SI
- Published
- 2019
48. Correction: The necroptosis machinery mediates axonal degeneration in a model of Parkinson disease
- Author
-
Maritza Oñate, Felipe A. Court, Alexis Martinez, Ines Moreno-Gonzalez, Claudio Soto, Nazaret Gamez, Claudio Hetz, Alejandra Catenaccio, Cristian Saquel, Natalia Salvadores, and Paulina Soto
- Subjects
Necroptosis ,Cell Biology ,Disease ,Biology ,Molecular Biology ,Axonal degeneration ,Neuroscience - Published
- 2020
- Full Text
- View/download PDF
49. Generation of a humanized Aβ expressing mouse demonstrating aspects of Alzheimer's disease-like pathology
- Author
-
Alessandra C. Martini, Kristine Minh Tran, Kelly Do Huynh, Cristina Nuñez-Diaz, Franklin Garcia, Celia da Cunha, Kim N. Green, Andrea J. Tenner, Rahasson R. Ager, Stefania Forner, David Baglietto-Vargas, Dina P. Matheos, Marcelo A. Wood, Carlos J. Rodriguez-Ortiz, Laura Trujillo-Estrada, Frank M. LaFerla, Enikö A. Kramár, Jimmy Phan, Xinyi Ma, Claudio Soto, Ines Moreno-Gonzalez, Grant R. MacGregor, Vivek Swarup, Juan Antonio García-León, Lena Cai, Masashi Kitazawa, Dominic I. Javonillo, Mohammad Shahnawaz, Jessica E. Childs, Antonia Gutierrez, Ali Mortazavi, Shan Jiang, Marie Minh Thu Nguyen, Gabriela Balderrama-Gutierrez, Ken C. Walls, [Baglietto-Vargas,D, Forner,S, Cai,L, Martini,AC, Trujillo-Estrada,L, Swarup,V, Nguyen,MMT, Do Huynh,K, Javonillo,DI, Tran,KM, Phan,J, Kramár,EA, Garcia,F, Childs,J, Rodriguez-Ortiz,CJ, Kitazawa,M, Matheos,DP, Da Cunha,C, Walls,KC, Ager,RR, Tenner,AJ, Wood,M, Green,KM, LaFerla,FM] Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA. [Baglietto-Vargas,D, LaFerla,FM] Department of Neurobiology and Behavior, University of California, Irvine, CA, USA. [Baglietto-Vargas,D, Nuñez-Diaz,C, Garcia-Leon,JA, Gutierrez,A, Moreno-Gonzalez,I] Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, Instituto de Investigacion Biomedica de Malaga-IBIMA, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Malaga, Malaga, Spain. [Jiang,S, Balderrama-Gutierrez,G, Ma,X, Mortazavi,A, MacGregor,GR] Department of Developmental and Cell Biology, University of California, Irvine, CA, USA. [Rodriguez-Ortiz,CJ, Kitazawa,M] Division of Occupational and Environmental Medicine, Department of Medicine. Center for Occupational and Environmental Health (COEH), University of California, Irvine, CA, USA. [Shahnawaz,M, Soto,C, Moreno-Gonzale,I] The Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA. [Tenner,AJ] Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA., and This study was initiated by the generosity of Harry Bubb through Cure Alzheimer’s Fund CAF-50997 (F.M.L.). Additional support was from Alzheimer’s Association NIRG-15-363477 (D.B.V.), AARF-16-440760 (S.F.) and NIRG-394284 (I.M.G.), The Larry Hillblom Foundation 2013-A-016-FEL (D.B.V.) and 2016-A-016-FEL (A.C.M.), the National Institute of Health (NIH) NIH/NIA/NINDS AG027544 (F.M.L.), AG00538 (F.M.L.), AG54884 (F.M.L.), OD010420 (F.M.L.), U54 AG054349 (F.M.L., A.J.T.), AG049562 (C.S.) NS083801 (K.N.G.) and AG056768 (K.N.G.), BrightFocus Foundation grant A2015535S (F.M.L.), by Minister of Science and Innovation grant PID2019-108911RA-100 (D.B.V.), Beatriz Galindo program BAGAL18/00052 (D.B.V.) and Institute of Health Carlos III (ISCiii) grant PI18/01557 (A.G.) co-financed by FEDER funds from European Union, by American federation of aging research-AFAR young investigator award and UC Irvine startup funds (V.S.) and UCI MIND pilot project (D.B.V.). The UCI-ADRC is funded by NIH/NIA Grant P50 AG16573 (F.M.L.). Genetically modified hAβ-KI mice were generated by the UCI Transgenic Mouse Facility, a shared resource funded in part by the Chao Family Comprehensive Cancer Center Support Grant (P30CA062203) from the National Cancer Institute. We thank Drs. Malcolm Leissring and Rodrigo Medeiros for critically reading the manuscript.
- Subjects
0301 basic medicine ,Male ,Aging ,Disciplines and Occupations::Natural Science Disciplines::Biological Science Disciplines::Biology::Genetics::Genetic Research::Gene Ontology [Medical Subject Headings] ,Information Science::Information Science::Computing Methodologies::Software::Mobile Applications [Medical Subject Headings] ,General Physics and Astronomy ,Neurodegenerative ,medicine.disease_cause ,Inbred C57BL ,Alzheimer's Disease ,Phenomena and Processes::Genetic Phenomena::Genetic Structures::Genome::Genome Components::Genes::Gene Components::Exons [Medical Subject Headings] ,Phenomena and Processes::Genetic Phenomena::Genetic Processes::Gene Expression [Medical Subject Headings] ,Transgenic ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Primates::Haplorhini::Catarrhini::Hominidae::Humans [Medical Subject Headings] ,Exon ,Mice ,Amyloid beta-Protein Precursor ,Cognition ,0302 clinical medicine ,Enfermedad de Alzheimer ,Gene expression ,Organisms::Eukaryota::Animals [Medical Subject Headings] ,2.1 Biological and endogenous factors ,Gene Regulatory Networks ,Organisms::Eukaryota::Animals::Animal Population Groups::Animals, Genetically Modified::Mice, Transgenic [Medical Subject Headings] ,Aetiology ,Phenomena and Processes::Musculoskeletal and Neural Physiological Phenomena::Nervous System Physiological Phenomena::Nervous System Physiological Processes::Neuronal Plasticity [Medical Subject Headings] ,Mutation ,Multidisciplinary ,Neuronal Plasticity ,Reacción del ácido peryódico de Schiff ,Brain ,Alzheimer's disease ,Cell biology ,Chemicals and Drugs::Inorganic Chemicals::Acids::Acids, Noncarboxylic::Periodic Acid [Medical Subject Headings] ,Neurological ,Female ,Diseases::Nervous System Diseases::Neurodegenerative Diseases::Tauopathies::Alzheimer Disease [Medical Subject Headings] ,Phenomena and Processes::Genetic Phenomena::Genetic Variation::Mutation [Medical Subject Headings] ,Periodic acid-schiff ,Science ,Transgene ,Check Tags::Male [Medical Subject Headings] ,Cre recombinase ,Phenomena and Processes::Genetic Phenomena::Genetic Structures::Base Sequence::Regulatory Sequences, Nucleic Acid::Gene Regulatory Networks [Medical Subject Headings] ,Locus (genetics) ,Mice, Transgenic ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Investigative Techniques::Genetic Techniques::Gene Expression Profiling [Medical Subject Headings] ,Biology ,Molecular neuroscience ,Exones ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Alzheimer Disease ,medicine ,Acquired Cognitive Impairment ,Genetics ,Animals ,Humans ,Chemicals and Drugs::Amino Acids, Peptides, and Proteins::Amino Acids [Medical Subject Headings] ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Rodentia::Muridae::Murinae::Mice [Medical Subject Headings] ,Cognición ,Psychiatry and Psychology::Psychological Phenomena and Processes::Mental Processes::Cognition [Medical Subject Headings] ,Amyloid beta-Peptides ,Mutación ,Animal ,Gene Expression Profiling ,Wild type ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Chemicals and Drugs::Amino Acids, Peptides, and Proteins::Peptides::Amyloid beta-Peptides [Medical Subject Headings] ,Anatomy::Nervous System::Central Nervous System::Brain [Medical Subject Headings] ,General Chemistry ,Chemicals and Drugs::Amino Acids, Peptides, and Proteins::Proteins::Amyloid::Amyloidogenic Proteins::Amyloid beta-Protein Precursor [Medical Subject Headings] ,Diseases::Animal Diseases::Disease Models, Animal [Medical Subject Headings] ,Brain Disorders ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Gene Ontology ,Check Tags::Female [Medical Subject Headings] ,Synaptic plasticity ,Disease Models ,Dementia ,Organisms::Eukaryota::Animals::Animal Population Groups::Animals, Laboratory::Animals, Inbred Strains::Mice, Inbred Strains::Mice, Inbred C57BL [Medical Subject Headings] ,030217 neurology & neurosurgery ,Expresión génica - Abstract
The majority of Alzheimer’s disease (AD) cases are late-onset and occur sporadically, however most mouse models of the disease harbor pathogenic mutations, rendering them better representations of familial autosomal-dominant forms of the disease. Here, we generated knock-in mice that express wildtype human Aβ under control of the mouse App locus. Remarkably, changing 3 amino acids in the mouse Aβ sequence to its wild-type human counterpart leads to age-dependent impairments in cognition and synaptic plasticity, brain volumetric changes, inflammatory alterations, the appearance of Periodic Acid-Schiff (PAS) granules and changes in gene expression. In addition, when exon 14 encoding the Aβ sequence was flanked by loxP sites we show that Cre-mediated excision of exon 14 ablates hAβ expression, rescues cognition and reduces the formation of PAS granules., Most instances of Alzheimer’s disease (AD) are sporadic or not associated with a particular mutation. Here, the authors develop knock-in mice that express wildtype human Aβ under control of the mouse App locus, which may have potential for modelling some aspects of sporadic late onset AD.
- Published
- 2018
50. Soft-Lithographic Patterning of Luminescent Carbon Nanodots Derived from Collagen Waste
- Author
-
Sandhya Susarla, Angel A. Martí, Valery N. Khabashesku, Amir Aliyan, Sadegh Yazdi, Ashokkumar Meiyazhagan, Anumary Ayyappan, Pulickel M. Ajayan, Robert Vajtai, Ines Moreno-Gonzalez, and Karina Cuanalo-Contreras
- Subjects
Photoluminescence ,Materials science ,Luminescence ,Biocompatibility ,Nanophotonics ,Nanotechnology ,Biocompatible Materials ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Fluorescence ,Mice ,Carbon nanodots ,Quantum Dots ,Spectroscopy, Fourier Transform Infrared ,Nanobiotechnology ,Animals ,General Materials Science ,Lithography ,Photoelectron Spectroscopy ,Water ,021001 nanoscience & nanotechnology ,Carbon ,0104 chemical sciences ,Quantum dot ,Printing ,Collagen ,0210 nano-technology - Abstract
Luminescent carbon dots (Cdots) synthesized using inexpensive precursors have inspired tremendous research interest because of their superior properties and applicability in various fields. In this work, we report a simple, economical, green route for the synthesis of multifunctional fluorescent Cdots prepared from a natural, low-cost source: collagen extracted from animal skin wastes. The as-synthesized metal-free Cdots were found to be in the size range of ∼1.2-9 nm, emitting bright blue photoluminescence with a calculated Cdot yield of ∼63%. Importantly, the soft-lithographic method used was inexpensive and yielded a variety of Cdot patterns with different geometrical structures and significant cellular biocompatibility. This novel approach to Cdot production highlights innovative ways of transforming industrial biowastes into advanced multifunctional materials which offer exciting potential for applications in nanophotonics and nanobiotechnology using a simple and scalable technique.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.