Network


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

Hotspot


Dive into the research topics where Phillip D. Rivera is active.

Publication


Featured researches published by Phillip D. Rivera.


The Journal of Neuroscience | 2010

Notch1 Is Required for Maintenance of the Reservoir of Adult Hippocampal Stem Cells

Jessica L. Ables; Nathan A. DeCarolis; Madeleine A. Johnson; Phillip D. Rivera; Zhengliang Gao; Donald C. Cooper; Freddy Radtke; Jenny Hsieh; Amelia J. Eisch

Notch1 regulates neural stem cell (NSC) number during development, but its role in adult neurogenesis is unclear. We generated nestin-CreERT2/R26R-YFP/Notch1loxP/loxP [Notch1inducible knock-out (iKO)] mice to allow tamoxifen (TAM)-inducible elimination of Notch1 and concomitant expression of yellow fluorescent protein (YFP) in nestin-expressing Type-1 NSCs and their progeny in the adult hippocampal subgranular zone (SGZ). Consistent with previous research, YFP+ cells in all stages of neurogenesis were evident in the subgranular zone (SGZ) of wild-type (WT) mice (nestin-CreERT2/R26R-YFP/Notch1w/w) after tamoxifen (post-TAM), producing adult-generated YFP+ dentate gyrus neurons. Compared with WT littermates, Notch1 iKO mice had similar numbers of total SGZ YFP+ cells 13 and 30 d post-TAM but had significantly fewer SGZ YFP+ cells 60 and 90 d post-TAM. Significantly fewer YFP+ Type-1 NSCs and transiently amplifying progenitors (TAPs) resulted in generation of fewer YFP+ granule neurons in Notch1 iKO mice. Strikingly, 30 d of running rescued this deficit, as the total YFP+ cell number in Notch iKO mice was equivalent to WT levels. This was even more notable given the persistent deficits in the Type-1 NSC and TAP reservoirs. Our data show that Notch1 signaling is required to maintain a reservoir of undifferentiated cells and ensure continuity of adult hippocampal neurogenesis, but that alternative Notch- and Type-1 NSC-independent pathways compensate in response to physical activity. These data shed light on the complex relationship between Type-1 NSCs, adult neurogenesis, the neurogenic niche, and environmental stimuli.


Molecular Psychiatry | 2015

The P7C3 class of neuroprotective compounds exerts antidepressant efficacy in mice by increasing hippocampal neurogenesis

Angela K. Walker; Phillip D. Rivera; Qian Wang; Jen-Chieh Chuang; Stephanie Tran; Sherri Osborne-Lawrence; Sandi Jo Estill; Ruth Starwalt; Paula Huntington; Lorraine K. Morlock; Jacinth Naidoo; Noelle S. Williams; Joseph M. Ready; Amelia J. Eisch; Andrew A. Pieper; Jeffrey M. Zigman

Augmenting hippocampal neurogenesis represents a potential new strategy for treating depression. Here we test this possibility by comparing hippocampal neurogenesis in depression-prone ghrelin receptor (Ghsr)-null mice to that in wild-type littermates and by determining the antidepressant efficacy of the P7C3 class of neuroprotective compounds. Exposure of Ghsr-null mice to chronic social defeat stress (CSDS) elicits more severe depressive-like behavior than in CSDS-exposed wild-type littermates, and exposure of Ghsr-null mice to 60% caloric restriction fails to elicit antidepressant-like behavior. CSDS resulted in more severely reduced cell proliferation and survival in the ventral dentate gyrus (DG) subgranular zone of Ghsr-null mice than in that of wild-type littermates. Also, caloric restriction increased apoptosis of DG subgranular zone cells in Ghsr-null mice, although it had the opposite effect in wild-type littermates. Systemic treatment with P7C3 during CSDS increased survival of proliferating DG cells, which ultimately developed into mature (NeuN+) neurons. Notably, P7C3 exerted a potent antidepressant-like effect in Ghsr-null mice exposed to either CSDS or caloric restriction, while the more highly active analog P7C3-A20 also exerted an antidepressant-like effect in wild-type littermates. Focal ablation of hippocampal stem cells with radiation eliminated this antidepressant effect, further attributing the P7C3 class antidepressant effect to its neuroprotective properties and resultant augmentation of hippocampal neurogenesis. Finally, P7C3-A20 demonstrated greater proneurogenic efficacy than a wide spectrum of currently marketed antidepressant drugs. Taken together, our data confirm the role of aberrant hippocampal neurogenesis in the etiology of depression and suggest that the neuroprotective P7C3-compounds represent a novel strategy for treating patients with this disease.


Neuropsychopharmacology | 2017

Glial and Neuroimmune Mechanisms as Critical Modulators of Drug Use and Abuse.

Michael J. Lacagnina; Phillip D. Rivera; Staci D. Bilbo

Drugs of abuse cause persistent alterations in synaptic plasticity that may underlie addiction behaviors. Evidence suggests glial cells have an essential and underappreciated role in the development and maintenance of drug abuse by influencing neuronal and synaptic functions in multifaceted ways. Microglia and astrocytes perform critical functions in synapse formation and refinement in the developing brain, and there is growing evidence that disruptions in glial function may be implicated in numerous neurological disorders throughout the lifespan. Linking evidence of function in health and under pathological conditions, this review will outline the glial and neuroimmune mechanisms that may contribute to drug-abuse liability, exploring evidence from opioids, alcohol, and psychostimulants. Drugs of abuse can activate microglia and astrocytes through signaling at innate immune receptors, which in turn influence neuronal function not only through secretion of soluble factors (eg, cytokines and chemokines) but also potentially through direct remodeling of the synapses. In sum, this review will argue that neural–glial interactions represent an important avenue for advancing our understanding of substance abuse disorders.


Radiation Research | 2013

Acute and Fractionated Exposure to High-LET 56Fe HZE-Particle Radiation Both Result in Similar Long-Term Deficits in Adult Hippocampal Neurogenesis

Phillip D. Rivera; Hung Ying Shih; Junie A. Leblanc; Mara G. Cole; Wellington Z. Amaral; Shibani Mukherjee; Shichuan Zhang; Melanie J. Lucero; Nathan A. DeCarolis; Benjamin P C Chen; Amelia J. Eisch

Astronauts on multi-year interplanetary missions will be exposed to a low, chronic dose of high-energy, high-charge particles. Studies in rodents show acute, nonfractionated exposure to these particles causes brain changes such as fewer adult-generated hippocampal neurons and stem cells that may be detrimental to cognition and mood regulation and thus compromise mission success. However, the influence of a low, chronic dose of these particles on neurogenesis and stem cells is unknown. To examine the influence of galactic cosmic radiation on neurogenesis, adult-generated stem and progenitor cells in Nestin-CreERT2/R26R-YFP transgenic mice were inducibly labeled to allow fate tracking. Mice were then sham exposed or given one acute 100 cGy 56Fe-particle exposure or five fractionated 20 cGy 56Fe-particle exposures. Adult-generated hippocampal neurons and stem cells were quantified 24 h or 3 months later. Both acute and fractionated exposure decreased the amount of proliferating cells and immature neurons relative to sham exposure. Unexpectedly, neither acute nor fractionated exposure decreased the number of adult neural stem cells relative to sham expsoure. Our findings show that single and fractionated exposures of 56Fe-particle irradiation are similarly detrimental to adult-generated neurons. Implications for future missions and ground-based studies in space radiation are discussed.


Neuroscience Letters | 2015

Chronic P7C3 treatment restores hippocampal neurogenesis

Sarah E. Latchney; Thomas C. Jaramillo; Phillip D. Rivera; Amelia J. Eisch; Craig M. Powell

Down syndrome (DS) is the most common genetic cause of intellectual disability and developmental delay. In addition to cognitive dysfunction, DS patients are marked by diminished neurogenesis, a neuropathological feature also found in the Ts65Dn mouse model of DS. Interestingly, manipulations that enhance neurogenesis - like environmental enrichment or pharmacological agents - improve cognition in Ts65Dn mice. P7C3 is a proneurogenic compound that enhances hippocampal neurogenesis, cell survival, and promotes cognition in aged animals. However, this compound has not been tested in the Ts65Dn mouse model of DS. We hypothesized that P7C3 treatment would reverse or ameliorate the neurogenic deficits in Ts65Dn mice. To test this, adult Ts65Dn and age-matched wild-type (WT) mice were administered vehicle or P7C3 twice daily for 3 months. After 3 months, brains were examined for indices of neurogenesis, including quantification of Ki67, DCX, activated caspase-3 (AC3), and surviving BrdU-immunoreactive(+) cells in the granule cell layer (GCL) of the hippocampal dentate gyrus. P7C3 had no effect on total Ki67+, DCX+, AC3+, or surviving BrdU+ cells in WT mice relative to vehicle. GCL volume was also not changed. In keeping with our hypothesis, however, P7C3-treated Ts65Dn mice had a significant increase in total Ki67+, DCX+, and surviving BrdU+ cells relative to vehicle. P7C3 treatment also decreased AC3+ cell number but had no effect on total GCL volume in Ts65Dn mice. Our findings show 3 months of P7C3 is sufficient to restore the neurogenic deficits observed in the Ts65Dn mouse model of DS.


Journal of Applied Physiology | 2014

The effect of spaceflight on mouse olfactory bulb volume, neurogenesis, and cell death indicates the protective effect of novel environment

Sarah E. Latchney; Phillip D. Rivera; Xiao Wen Mao; Virginia L. Ferguson; Ted A. Bateman; Louis S. Stodieck; Gregory A. Nelson; Amelia J. Eisch

Space missions necessitate physiological and psychological adaptations to environmental factors not present on Earth, some of which present significant risks for the central nervous system (CNS) of crewmembers. One CNS region of interest is the adult olfactory bulb (OB), as OB structure and function are sensitive to environmental- and experience-induced regulation. It is currently unknown how the OB is altered by spaceflight. In this study, we evaluated OB volume and neurogenesis in mice shortly after a 13-day flight on Space Shuttle Atlantis [Space Transport System (STS)-135] relative to two groups of control mice maintained on Earth. Mice housed on Earth in animal enclosure modules that mimicked the conditions onboard STS-135 (AEM-Ground mice) had greater OB volume relative to mice maintained in standard housing on Earth (Vivarium mice), particularly in the granule (GCL) and glomerular (GL) cell layers. AEM-Ground mice also had more OB neuroblasts and fewer apoptotic cells relative to Vivarium mice. However, the AEM-induced increase in OB volume and neurogenesis was not seen in STS-135 mice (AEM-Flight mice), suggesting that spaceflight may have negated the positive effects of the AEM. In fact, when OB volume of AEM-Flight mice was considered, there was a greater density of apoptotic cells relative to AEM-Ground mice. Our findings suggest that factors present during spaceflight have opposing effects on OB size and neurogenesis, and provide insight into potential strategies to preserve OB structure and function during future space missions.


Hippocampus | 2015

Retrieval of morphine‐associated context induces cFos in dentate gyrus neurons

Phillip D. Rivera; Ramya K. Raghavan; Sanghee Yun; Sarah E. Latchney; Mary Katherin Mcgovern; Emily F. García; Shari G. Birnbaum; Amelia J. Eisch

Addiction has been proposed to emerge from associations between the drug and the reward‐associated contexts. This associative learning has a cellular correlate, as there are more cFos+ neurons in the hippocampal dentate gyrus (DG) after psychostimulant conditioned place preference (CPP) versus saline controls. However, it is unknown whether morphine CPP leads to a similar DG activation, or whether DG activation is due to locomotion, handling, pharmacological effects, or—as data from contextual fear learning suggests—exposure to the drug‐associated context. To explore this, we employed an unbiased, counterbalanced, and shortened CPP design that led to place preference and more DG cFos+ cells. Next, mice underwent morphine CPP but were then sequestered into the morphine‐paired (conditioned stimulus+ [CS+]) or saline‐paired (CS−) context on test day. Morphine‐paired mice sequestered to CS+ had ∼30% more DG cFos+ cells than saline‐paired mice. Furthermore, Bregma analysis revealed morphine‐paired mice had more cFos+ cells in CS+ compared to CS− controls. Notably, there was no significant difference in DG cFos+ cell number after handling alone or after receiving morphine in home cage. Thus, retrieval of morphine‐associated context is accompanied by activation of hippocampal DG granule cell neurons.


Journal of Neuroscience Methods | 2017

Optimized solubilization of TRIzol-precipitated protein permits Western blotting analysis to maximize data available from brain tissue

Ashley M. Kopec; Phillip D. Rivera; Michael J. Lacagnina; Richa Hanamsagar; Staci D. Bilbo

BACKGROUND Techniques simultaneously assessing multiple levels of molecular processing are appealing because molecular signaling underlying complex neural phenomena occurs at complementary levels. The TRIzol method isolates RNA and DNA, but protein retrieval is difficult due to inefficient solubilization of precipitated protein pellets. NEW METHOD We optimized a buffer for the efficient solubilization of protein from TRIzol-precipitated brain tissue for Western blotting analysis, which was also more effective at directly homogenizing brain tissue than RIPA buffer. RESULTS Protein yield during solubilization, in addition to protein yield via direct homogenization, is increased by optimizing concentrations of chemicals in a standard lysis buffer. Effective incubation parameters for both total protein yield and the analysis of post-translational modifications is remarkably flexible. Importantly, different neural cell types and protein classes are represented in solubilized protein samples. Moreover, we used dissociated mouse brain tissue to isolate microglia from other cell types and successfully resolved cell type-specific proteins from these small and difficult to attain samples. COMPARISON WITH EXISTING METHOD(S) Solubilization buffers to date have been comprised primarily of SDS or urea; the data herein demonstrate that components common to lysis buffers can also enhance protein solubilization both after direct homogenization and after precipitation. CONCLUSIONS This method is suitable for assessing gene and protein expression from a single brain sample, allowing for a more comprehensive evaluation of neural phenomena while minimizing the number of subjects.


Nature Medicine | 2018

Stimulation of entorhinal cortex–dentate gyrus circuitry is antidepressive

Sanghee Yun; Ryan P. Reynolds; Iraklis Petrof; Alicia White; Phillip D. Rivera; Amir Segev; Adam D. Gibson; Maiko Suarez; Matthew J. DeSalle; Naoki Ito; Shibani Mukherjee; Devon R. Richardson; Catherine E. Kang; Rebecca C. Ahrens-Nicklas; Ivan Soler; Dane M. Chetkovich; Saïd Kourrich; Douglas A. Coulter; Amelia J. Eisch

Major depressive disorder (MDD) is considered a ‘circuitopathy’, and brain stimulation therapies hold promise for ameliorating MDD symptoms, including hippocampal dysfunction. It is unknown whether stimulation of upstream hippocampal circuitry, such as the entorhinal cortex (Ent), is antidepressive, although Ent stimulation improves learning and memory in mice and humans. Here we show that molecular targeting (Ent-specific knockdown of a psychosocial stress-induced protein) and chemogenetic stimulation of Ent neurons induce antidepressive-like effects in mice. Mechanistically, we show that Ent-stimulation-induced antidepressive-like behavior relies on the generation of new hippocampal neurons. Thus, controlled stimulation of Ent hippocampal afferents is antidepressive via increased hippocampal neurogenesis. These findings emphasize the power and potential of Ent glutamatergic afferent stimulation—previously well-known for its ability to influence learning and memory—for MDD treatment.In mouse models of stress-induced depression, molecular and chemogenetic stimulation of the entorhinal cortex induces the production of adult-born hippocampal neurons and generates antidepressive-like effects.


Nature Medicine | 2018

Publisher Correction: Stimulation of entorhinal cortex–dentate gyrus circuitry is antidepressive

Sanghee Yun; Ryan P. Reynolds; Iraklis Petrof; Alicia White; Phillip D. Rivera; Amir Segev; Adam D. Gibson; Maiko Suarez; Matthew J. DeSalle; Naoki Ito; Shibani Mukherjee; Devon R. Richardson; Catherine E. Kang; Rebecca C. Ahrens-Nicklas; Ivan Soler; Dane M. Chetkovich; Saïd Kourrich; Douglas A. Coulter; Amelia J. Eisch

In the version of this article originally published, a URL provided in the Methods section was incorrect. The URL had a solidus at the end but should have appeared as http://www.nature.com/authors/policies/image.html. The error has been corrected in the PDF and HTML versions of this article.

Collaboration


Dive into the Phillip D. Rivera's collaboration.

Top Co-Authors

Avatar

Amelia J. Eisch

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sarah E. Latchney

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sanghee Yun

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Benjamin P C Chen

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Junie A. Leblanc

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Louis S. Stodieck

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Matthew J. DeSalle

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Nathan A. DeCarolis

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ryan P. Reynolds

Children's Hospital of Philadelphia

View shared research outputs
Researchain Logo
Decentralizing Knowledge