Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Héctor De Jesús-Cortés is active.

Publication


Featured researches published by Héctor De Jesús-Cortés.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Neuroprotective efficacy of aminopropyl carbazoles in a mouse model of Parkinson disease

Héctor De Jesús-Cortés; Pin Xu; Jordan Drawbridge; Sandi Jo Estill; Paula Huntington; Stephanie Tran; Jeremiah K. Britt; Rachel Tesla; Lorraine K. Morlock; Jacinth Naidoo; Lisa Melito; Gelin Wang; Noelle S. Williams; Joseph M. Ready; Steven L. McKnight; Andrew A. Pieper

We previously reported the discovery of P7C3, an aminopropyl carbazole having proneurogenic and neuroprotective properties in newborn neural precursor cells of the dentate gyrus. Here, we provide evidence that P7C3 also protects mature neurons in brain regions outside of the hippocampus. P7C3 blocks 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-mediated cell death of dopaminergic neurons in the substantia nigra of adult mice, a model of Parkinson disease (PD). Dose–response studies show that the P7C3 analog P7C3A20 blocks cell death with even greater potency and efficacy, which parallels the relative potency and efficacy of these agents in blocking apoptosis of newborn neural precursor cells of the dentate gyrus. P7C3 and P7C3A20 display similar relative effects in blocking 1-methyl-4-phenylpyridinium (MPP+)-mediated death of dopaminergic neurons in Caenorhabditis elegans, as well as in preserving C. elegans mobility following MPP+ exposure. Dimebon, an antihistaminergic drug that is weakly proneurogenic and neuroprotective in the dentate gyrus, confers no protection in either the mouse or the worm models of PD. We further demonstrate that the hippocampal proneurogenic efficacy of eight additional analogs of P7C3 correlates with their protective efficacy in MPTP-mediated neurotoxicity. In vivo screening of P7C3 analogs for proneurogenic efficacy in the hippocampus may thus provide a reliable means of predicting neuroprotective efficacy. We propose that the chemical scaffold represented by P7C3 and P7C3A20 provides a basis for optimizing and advancing pharmacologic agents for the treatment of patients with PD.


Nature | 2015

Wild-type microglia do not reverse pathology in mouse models of Rett syndrome.

Jieqi Wang; Jan Eike Wegener; Teng Wei Huang; Smitha Sripathy; Héctor De Jesús-Cortés; Pin Xu; Stephanie Tran; Whitney Knobbe; Vid Leko; Jeremiah K. Britt; Ruth Starwalt; Latisha McDaniel; Christopher S. Ward; Diana Parra; Benjamin Newcomb; Uyen Lao; Cynthia Nourigat; David Flowers; Sean M. Cullen; Nikolas L. Jorstad; Yue Yang; Lena Glaskova; Sebastian Vigneau; Julia Kozlitina; Michael J. Yetman; Joanna L. Jankowsky; Sybille D. Reichardt; Holger M. Reichardt; Jutta Gärtner; Marisa S. Bartolomei

arising from N. C. Derecki et al. 484, 105–109 (2012); doi:10.1038/nature10907Rett syndrome is a severe neurodevelopmental disorder caused by mutations in the X chromosomal gene MECP2 (ref. 1), and its treatment so far is symptomatic. Mecp2 disruption in mice phenocopies major features of the syndrome that can be reversed after Mecp2 re-expression. Recently, Derecki et al. reported that transplantation of wild-type bone marrow into lethally irradiated Mecp2-null (Mecp2tm1.1Jae/y) mice prevented neurological decline and early death by restoring microglial phagocytic activity against apoptotic targets, and clinical trials of bone marrow transplantation (BMT) for patients with Rett syndrome have thus been initiated. We aimed to replicate and extend the BMT experiments in three different Rett syndrome mouse models, but found that despite robust microglial engraftment, BMT from wild-type donors did not prevent early death or ameliorate neurological deficits. Furthermore, early and specific Mecp2 genetic expression in microglia did not rescue Mecp2-deficient mice.


Cell Reports | 2014

P7C3 Neuroprotective Chemicals Block Axonal Degeneration and Preserve Function after Traumatic Brain Injury

Terry C. Yin; Jeremiah K. Britt; Héctor De Jesús-Cortés; Yuan Lu; Rachel M. Genova; Michael Z. Khan; Jaymie R. Voorhees; Jianqiang Shao; Aaron Katzman; Paula Huntington; Cassie Wassink; Latisha McDaniel; Elizabeth A. Newell; Laura M. Dutca; Jacinth Naidoo; Huxing Cui; Alexander G. Bassuk; Matthew M. Harper; Steven L. McKnight; Joseph M. Ready; Andrew A. Pieper

The P7C3 class of neuroprotective aminopropyl carbazoles has been shown to block neuronal cell death in models of neurodegeneration. We now show that P7C3 molecules additionally preserve axonal integrity after injury, before neuronal cell death occurs, in a rodent model of blast-mediated traumatic brain injury (TBI). This protective quality may be linked to the ability of P7C3 molecules to activate nicotinamide phosphoribosyltransferase, the rate-limiting enzyme in nicotinamide adenine dinucleotide salvage. Initiation of daily treatment with our recently reported lead agent, P7C3-S243, 1 day after blast-mediated TBI blocks axonal degeneration and preserves normal synaptic activity, learning and memory, and motor coordination in mice. We additionally report persistent neurologic deficits and acquisition of an anxiety-like phenotype in untreated animals 8 months after blast exposure. Optimized variants of P7C3 thus offer hope for identifying neuroprotective agents for conditions involving axonal damage, neuronal cell death, or both, such as occurs in TBI.


Journal of Medicinal Chemistry | 2014

Discovery of a Neuroprotective Chemical, (S)-N-(3-(3,6-Dibromo-9H-carbazol-9-yl)-2-fluoropropyl)-6-methoxypyridin-2-amine [(−)-P7C3-S243], with Improved Druglike Properties

Jacinth Naidoo; Héctor De Jesús-Cortés; Paula Huntington; Sandi Jo Estill; Lorraine K. Morlock; Ruth Starwalt; Thomas J. Mangano; Noelle S. Williams; Andrew A. Pieper; Joseph M. Ready

(−)-P7C3-S243 is a neuroprotective aminopropyl carbazole with improved druglike properties compared with previously reported compounds in the P7C3 class. It protects developing neurons in a mouse model of hippocampal neurogenesis and protects mature neurons within the substantia nigra in a mouse model of Parkinson’s disease. A short, enantioselective synthesis provides the neuroprotective agent in optically pure form. It is nontoxic, orally bioavailable, metabolically stable, and able to cross the blood–brain barrier. As such, it represents a valuable lead compound for the development of drugs to treat neurodegenerative diseases and traumatic brain injury.


Journal of Neurochemistry | 2013

EFhd2 is a novel amyloid protein associated with pathological tau in Alzheimer's disease

Yancy Ferrer-Acosta; Eva N. Rodríguez-Cruz; François Orange; Héctor De Jesús-Cortés; Bismark Madera; Jaime Vaquer-Alicea; Juan Ballester; Maxime J.-F. Guinel; George S. Bloom; Irving E. Vega

EFhd2 is a conserved calcium‐binding protein, abundant within the central nervous system. Previous studies identified EFhd2 associated with pathological forms of tau proteins in the tauopathy mouse model JNPL3, which expresses the human tauP301L mutant. This association was validated in human tauopathies, such as Alzheimers disease (AD). However, the role that EFhd2 may play in tauopathies is still unknown. Here, we show that EFhd2 formed amyloid structures in vitro, a capability that is reduced by calcium ions. Electron microscopy (EM) analyses demonstrated that recombinant EFhd2 formed filamentous structures. EM analyses of sarkosyl‐insoluble fractions derived from human AD brains also indicated that EFhd2 co‐localizes with aggregated tau proteins and formed granular structures. Immunohistological analyses of brain slices demonstrated that EFhd2 co‐localizes with pathological tau proteins in AD brains, confirming the co‐aggregation of EFhd2 and pathological tau. Furthermore, EFhd2s coiled‐coil domain mediated its self‐oligomerization in vitro and its association with tau proteins in JNPL3 mouse brain extracts. The results demonstrate that EFhd2 is a novel amyloid protein associated with pathological tau proteins in AD brain and that calcium binding may regulate the formation of EFhd2s amyloid structures. Hence, EFhd2 may play an important role in the pathobiology of tau‐mediated neurodegeneration.


eNeuro | 2016

The Neuropsychiatric Disease-Associated Gene cacna1c Mediates Survival of Young Hippocampal Neurons.

Anni S. Lee; Héctor De Jesús-Cortés; Zeeba D. Kabir; Whitney Knobbe; Madeline Orr; Caitlin E. Burgdorf; Paula Huntington; Latisha McDaniel; Jeremiah K. Britt; Franz Hoffmann; Daniel J. Brat; Anjali M. Rajadhyaksha; Andrew A. Pieper

Visual Overview Genetic variations in CACNA1C, which encodes the Cav1.2 subunit of L-type calcium channels (LTCCs), are associated with multiple forms of neuropsychiatric disease that manifest high anxiety in patients. Genetic variations in CACNA1C, which encodes the Cav1.2 subunit of L-type calcium channels (LTCCs), are associated with multiple forms of neuropsychiatric disease that manifest high anxiety in patients. In parallel, mice harboring forebrain-specific conditional knockout of cacna1c (forebrain-Cav1.2 cKO) display unusually high anxiety-like behavior. LTCCs in general, including the Cav1.3 subunit, have been shown to mediate differentiation of neural precursor cells (NPCs). However, it has not previously been determined whether Cav1.2 affects postnatal hippocampal neurogenesis in vivo. Here, we show that forebrain-Cav1.2 cKO mice exhibit enhanced cell death of young hippocampal neurons, with no change in NPC proliferation, hippocampal size, dentate gyrus thickness, or corticosterone levels compared with wild-type littermates. These mice also exhibit deficits in brain levels of brain-derived neurotrophic factor (BDNF), and Cre recombinase-mediated knockdown of adult hippocampal Cav1.2 recapitulates the deficit in young hippocampal neurons survival. Treatment of forebrain-Cav1.2 cKO mice with the neuroprotective agent P7C3-A20 restored the net magnitude of postnatal hippocampal neurogenesis to wild-type levels without ameliorating their deficit in BDNF expression. The role of Cav1.2 in young hippocampal neurons survival may provide new approaches for understanding and treating neuropsychiatric disease associated with aberrations in CACNA1C. Visual Abstract


npj Parkinson's disease | 2015

Protective efficacy of P7C3-S243 in the 6-hydroxydopamine model of Parkinson's disease

Héctor De Jesús-Cortés; Adam D. Miller; Jeremiah K. Britt; Anthony J DeMarco; Mayralis De Jesús-Cortés; Emily Stuebing; Jacinth Naidoo; Edwin Vázquez-Rosa; Lorraine K. Morlock; Noelle S. Williams; Joseph M. Ready; Nandakumar S. Narayanan; Andrew A. Pieper

Background:There are currently no therapeutic options for patients with Parkinsons disease that prevent or slow the death of dopaminergic neurons. We have recently identified the novel P7C3 class of neuroprotective molecules that blocks neuron cell death.AIMS:The aim of this study was to determine whether treatment with highly active members of the P7C3 series blocks dopaminergic neuron cell death and associated behavioral and neurochemical deficits in the rat 6-hydroxydopamine (6-OHDA) model of Parkinsons disease.Methods:After unilateral injection of 6-OHDA into the median forebrain bundle, rats were assessed for behavioral function in the open field, cylinder test, and amphetamine-induced circling test. Thereafter, their brains were subjected to neurochemical and immunohistochemical analysis of dopaminergic neuron survival. Analysis was conducted as a function of treatment with P7C3 compounds, with administration initiated either before or after 6-OHDA exposure.Results:Animals administered P7C3-A20 or P7C3-S243, two of the most advanced agents in the P7C3 series of neuroprotective compounds, both before and after 6-OHDA exposure showed evidence of protective efficacy in all measures. When P7C3-S243 administration was initiated after 6-OHDA exposure, rats also showed protective efficacy in all measures, which included blocking dopaminergic neuron cell death in ipsilateral substantia nigra pars compacta, preservation of dopamine and its metabolites in ipsilateral striatum, and preservation of normal motor behavior.Conclusions:The P7C3 series of compounds may form the basis for developing new therapeutic agents for slowing or preventing progression of Parkinsons disease.


Neuroreport | 2012

Amphiphysin-1 protein level changes associated with tau-mediated neurodegeneration

Héctor De Jesús-Cortés; Carlos Nogueras-Ortiz; Marla Gearing; Steven E. Arnold; Irving E. Vega

Tauopathies are a family of neurodegenerative diseases that have the pathological hallmark of intraneuronal accumulation of filaments composed of hyperphosphorylated tau proteins that tend to aggregate in an ultrastructure known as neurofibrillary tangles. The identification of mutations on the tau gene in familial cases of tauopathies underscores the pathological role of the tau protein. However, the molecular process that underlines tau-mediated neurodegeneration is not understood. Here, a proteomics approach was used to identify proteins that may be affected during the course of tau-mediated neurodegeneration in the tauopathy mouse model JNPL3. The JNPL3 mice express human tau proteins bearing a P301L mutation, which mimics the neurodegenerative process observed in humans with tauopathy. The results showed that the protein amphiphysin-1 (AMPH1) is significantly reduced in terminally ill JNPL3 mice. Specifically, the AMPH1 protein level is reduced in brain regions known to accumulate aggregates of hyperphosphorylated tau proteins. The AMPH1 protein reduction was validated in Alzheimer’s disease cases. Taken together, the results suggest that the reduction of the AMPH1 protein level is a molecular event associated with the progression of tau-mediated neurodegeneration.


Brain Injury | 2018

Repeated mild traumatic brain injury produces neuroinflammation, anxiety-like behaviour and impaired spatial memory in mice

John I. Broussard; Laura Acion; Héctor De Jesús-Cortés; Terry Yin; Jeremiah K. Britt; Ramiro Salas; Mauro Costa-Mattioli; Claudia S. Robertson; Andrew A Pieper; David B. Arciniegas; Ricardo E. Jorge

ABSTRACT Primary Objective: Repeated traumatic brain injuries (rmTBI) are frequently associated with debilitating neuropsychiatric conditions such as cognitive impairment, mood disorders, and post-traumatic stress disorder. We tested the hypothesis that repeated mild traumatic brain injury impairs spatial memory and enhances anxiety-like behaviour. Research Design: We used a between groups design using single (smTBI) or repeated (rmTBI) controlled cranial closed skull impacts to mice, compared to a control group. Methods and Procedures: We assessed the effects of smTBI and rmTBI using measures of motor performance (Rotarod Test [RT]), anxiety-like behaviour (Elevated Plus Maze [EPM] and Open Field [OF] tests), and spatial memory (Morris Water Maze [MWM]) within 12 days of the final injury. In separate groups of mice, astrocytosis and microglial activation were assessed 24 hours after the final injury using GFAP and IBA-1 immunohistochemistry. Main Outcomes and Results: RmTBI impaired spatial memory in the MWM and increased anxiety-like behaviour in the EPM and OFT. In addition, rmTBI elevated GFAP and IBA-1 immunohistochemistry throughout the mouse brain. RmTBI produced astrocytosis and microglial activation, and elicited impaired spatial memory and anxiety-like behaviour. Conclusions: rmTBI produces acute cognitive and anxiety-like disturbances associated with inflammatory changes in brain regions involved in spatial memory and anxiety.


Frontiers in Neuroscience | 2014

Novel autoimmune response in a tauopathy mouse model

Carlos Nogueras-Ortiz; Héctor De Jesús-Cortés; Jaime Vaquer-Alicea; Irving E. Vega

Molecular diagnostic tools with non-invasive properties that allow detection of pathological events in Alzheimers disease (AD) and other neurodegenerative tauopathies are essential for the development of therapeutics. Several diagnostic strategies based on the identification of biomarkers have been proposed. However, its specificity among neurodegenerative disorders is disputable as the association with pathological events remains elusive. Recently, we showed that Amphiphysin-1 (AMPH1) proteins abundance is reduced in the central nervous system (CNS) of the tauopathy mouse model JNPL3 and AD brains. AMPH1 is a synaptic protein that plays an important role in clathrin-mediated endocytosis and associates with BIN1, one of the most important risk loci for AD. Also, it has been associated with a rare neurological disease known as Stiff-Person Syndrome (SPS). Auto-antibodies against AMPH1 are used as diagnostic biomarkers for a paraneoplastic variant of SPS. Therefore, we set up to evaluate the presence and abundance of auto-AMPH1 antibodies in tau-mediated neurodegeneration. Immunoblots and enzyme-linked immunosorbent assays (ELISA) were conducted to detect the presence of auto-AMPH1 antibodies in sera from euthanized mice that developed neurodegeneration (JNPL3) and healthy control mice (NTg). Results showed increased levels of auto-AMPH1 antibodies in JNPL3 sera compared to NTg controls. The abundance of auto-AMPH1 antibodies correlated with motor impairment and AMPH1 protein level decrease in the CNS. The results suggest that auto-AMPH1 antibodies could serve as a biomarker for the progression of tau-mediated neurodegeneration in JNPL3 mice.

Collaboration


Dive into the Héctor De Jesús-Cortés's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew A. Pieper

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Latisha McDaniel

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jieqi Wang

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Pin Xu

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Ruth Starwalt

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Stephanie Tran

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Whitney Knobbe

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge