Network


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

Hotspot


Dive into the research topics where Demian Obregon is active.

Publication


Featured researches published by Demian Obregon.


Journal of Neuroinflammation | 2005

Stimulation of cannabinoid receptor 2 (CB2) suppresses microglial activation.

Jared Ehrhart; Demian Obregon; Takashi Mori; Huayan Hou; Nan Sun; Yun Bai; Thomas W. Klein; Francisco Fernandez; Jun Tan; R. Douglas Shytle

BackgroundActivated microglial cells have been implicated in a number of neurodegenerative disorders, including Alzheimers disease (AD), multiple sclerosis (MS), and HIV dementia. It is well known that inflammatory mediators such as nitric oxide (NO), cytokines, and chemokines play an important role in microglial cell-associated neuron cell damage. Our previous studies have shown that CD40 signaling is involved in pathological activation of microglial cells. Many data reveal that cannabinoids mediate suppression of inflammation in vitro and in vivo through stimulation of cannabinoid receptor 2 (CB2).MethodsIn this study, we investigated the effects of a cannabinoid agonist on CD40 expression and function by cultured microglial cells activated by IFN-γ using RT-PCR, Western immunoblotting, flow cytometry, and anti-CB2 small interfering RNA (siRNA) analyses. Furthermore, we examined if the stimulation of CB2 could modulate the capacity of microglial cells to phagocytise Aβ1–42 peptide using a phagocytosis assay.ResultsWe found that the selective stimulation of cannabinoid receptor CB2 by JWH-015 suppressed IFN-γ-induced CD40 expression. In addition, this CB2 agonist markedly inhibited IFN-γ-induced phosphorylation of JAK/STAT1. Further, this stimulation was also able to suppress microglial TNF-α and nitric oxide production induced either by IFN-γ or Aβ peptide challenge in the presence of CD40 ligation. Finally, we showed that CB2 activation by JWH-015 markedly attenuated CD40-mediated inhibition of microglial phagocytosis of Aβ1–42 peptide. Taken together, these results provide mechanistic insight into beneficial effects provided by cannabinoid receptor CB2 modulation in neurodegenerative diseases, particularly AD.


Nature Neuroscience | 2002

Role of CD40 ligand in amyloidosis in transgenic Alzheimer's mice

Jun Tan; Terrence Town; Fiona Crawford; Takashi Mori; Anthony DelleDonne; Robert Crescentini; Demian Obregon; Richard A. Flavell; Michael Mullan

We have shown that interaction of CD40 with CD40L enables microglial activation in response to amyloid-β peptide (Aβ), which is associated with Alzheimers disease (AD)-like neuronal tau hyperphosphorylation in vivo. Here we report that transgenic mice overproducing Aβ, but deficient in CD40L, showed decreased astrocytosis and microgliosis associated with diminished Aβ levels and β-amyloid plaque load. Furthermore, in the PSAPP transgenic mouse model of AD, a depleting antibody against CD40L caused marked attenuation of Aβ/β-amyloid pathology, which was associated with decreased amyloidogenic processing of amyloid precursor protein (APP) and increased circulating levels of Aβ. Conversely, in neuroblastoma cells overexpressing wild-type human APP, the CD40–CD40L interaction resulted in amyloidogenic APP processing. These findings suggest several possible mechanisms underlying mitigation of AD pathology in response to CD40L depletion, and validate the CD40–CD40L interaction as a target for therapeutic intervention in AD.


Journal of Biological Chemistry | 2006

ADAM10 Activation Is Required for Green Tea (–)-Epigallocatechin-3-gallate-induced α-Secretase Cleavage of Amyloid Precursor Protein

Demian Obregon; Kavon Rezai-Zadeh; Yun Bai; Nan Sun; Huayan Hou; Jared Ehrhart; Jin Zeng; Takashi Mori; Gary W. Arendash; Doug Shytle; Terrence Town; Jun Tan

Recently, we have shown that green tea polyphenol (–)-epigallocatechin-3-gallate (EGCG) exerts a beneficial role on reducing brain Aβ levels, resulting in mitigation of cerebral amyloidosis in a mouse model of Alzheimer disease. EGCG seems to accomplish this by modulating amyloid precursor protein (APP) processing, resulting in enhanced cleavage of the α-COOH-terminal fragment (α-CTF) of APP and corresponding elevation of the NH2-terminal APP product, soluble APP-α (sAPP-α). These beneficial effects were associated with increased α-secretase cleavage activity, but no significant alteration in β-or γ-secretase activities. To gain insight into the molecular mechanism whereby EGCG modulates APP processing, we evaluated the involvement of three candidateα-secretase enzymes, a-disintegrin and metalloprotease (ADAM) 9, 10, or 17, in EGCG-induced non-amyloidogenic APP metabolism. Results show that EGCG treatment of N2a cells stably transfected with “Swedish” mutant human APP (SweAPP N2a cells) leads to markedly elevated active (∼60 kDa mature form) ADAM10 protein. Elevation of active ADAM10 correlates with increased α-CTF cleavage, and elevated sAPP-α. To specifically test the contribution of ADAM10 to non-amyloidogenic APP metabolism, small interfering RNA knockdown of ADAM9, -10, or -17 mRNA was employed. Results show that ADAM10 (but not ADAM9 or -17) is critical for EGCG-mediated α-secretase cleavage activity. In summary, ADAM10 activation is necessary for EGCG promotion of non-amyloidogenic (α-secretase cleavage) APP processing. Thus, ADAM10 represents an important pharmacotherapeutic target for the treatment of cerebral amyloidosis in Alzheimer disease.


The EMBO Journal | 2002

CD40 is expressed and functional on neuronal cells.

Jun Tan; Terrence Town; Takashi Mori; Demian Obregon; Yajuan Wu; Anthony DelleDonne; Amyn M. Rojiani; Fiona Crawford; Richard A. Flavell; Mike Mullan

We show here that CD40 mRNA and protein are expressed by neuronal cells, and are increased in differentiated versus undifferentiated N2a and PC12 cells as measured by RT–PCR, western blotting and immunofluorescence staining. Additionally, immunohistochemistry reveals that neurons from adult mouse and human brain also express CD40 in situ. CD40 ligation results in a time‐dependent increase in p44/42 MAPK activation in neuronal cells. Furthermore, ligation of CD40 opposes JNK phosphorylation and activity induced by NGF‐β removal from differentiated PC12 cells or serum withdrawal from primary cultured neurons. Importantly, CD40 ligation also protects neuronal cells from NGF‐β or serum withdrawal‐induced injury and affects neuronal differentiation. Finally, adult mice deficient for the CD40 receptor demonstrate neuronal dysfunction as evidenced by decreased neurofilament isoforms, reduced Bcl‐xL:Bax ratio, neuronal morphological change, increased DNA fragmentation, and gross brain abnormality. These changes occur with age, and are clearly evident at 16 months. Taken together, these data demonstrate a role of CD40 in neuronal development, maintenance and protection in vitro and in vivo.


Neuroscience Letters | 2001

Characterization of murine immunoglobulin G antibodies against human amyloid-β1–42

Terrence Town; Jun Tan; Nicholas Sansone; Demian Obregon; Thomas W. Klein; Michael Mullan

It has been demonstrated that immunization of transgenic mouse models of Alzheimers disease (AD) with amyloid-beta1-42 peptide (Abeta1-42) results in amelioration of AD-like pathology, including reduced soluble and deposited beta-amyloid and decreased cognitive impairment. Based on the proposed importance of immunoglobulin G (IgG) anti-Abeta antibodies (Abs) in these effects, we sought to characterize these Abs in splenocytes from mice immunized with Abeta1-42. Data show that a more aggregated preparation of Abeta1-42 gives a robust IgG anti-Abeta Ab response, while these Abs are almost undetectable when a less aggregated preparation of Abeta1-42 is used as the immunogen. Importantly, IgG anti-Abeta Ab production is detected after just 12 weeks of Abeta1-42 treatment. Analysis of anti-Abeta Ab IgG isotypes reveals that the majority of these Abs are IgG1, with significantly fewer Abs of the IgG2a or IgG2b isotypes (IgG1>IgG2a>IgG2b), suggesting a T lymphocyte helper type II response after Abeta1-42 immunization. To determine the epitope of Abeta recognized by IgG anti-Abeta Abs, intact Abeta and Abeta peptide fragments were analyzed for their ability to bind these Abs. Data show that these Abs specifically recognize an amino-terminal epitope of Abeta between amino acids one and twelve, with higher affinity for a more soluble preparation of Abeta1-42. These data further indicate the immunogenic potential of Abeta1-42 and offer insight into the nature of the IgG anti-Abeta Ab response.


Journal of Neuroscience Research | 2002

p35/Cdk5 pathway mediates soluble amyloid-β peptide-induced tau phosphorylation in vitro

Terrence Town; Joseph Zolton; Reed Shaffner; Billy Schnell; Robert Crescentini; Yajuan Wu; Jin Zeng; Anthony DelleDonne; Demian Obregon; Jun Tan; Mike Mullan

Alzheimers disease (AD) is pathologically characterized by deposition of amyloid‐β peptides (Aβ) as senile plaques and by the occurrence of neurofibrillary tangles (NFTs) composed primarily of hyperphosphorylated tau protein. Activation of cyclin‐dependent kinase 5 (Cdk5) via its potent activator p25 has recently been shown to promote phosphorylation of tau at AD‐specific phosphoepitopes, and increased cleavage of p35 to p25 has been demonstrated in AD patients, suggesting that Cdk5 may represent a pathogenic tau protein kinase. We were interested in the potential effect of soluble forms of Aβ on Cdk5‐mediated AD‐like tau phosphorylation, insofar as previous studies of human biopsies and aged canine and primate brains have shown that dystrophic neurites appear before the formation of neuritic plaques. We transfected N2a cells with a p35 vector (N2a/p35 cells) and, after differentiation, challenged these cells with Aβ1–42 peptide in soluble form (sAβ1–42). Results show that sAβ1–42 at relatively low levels (1–5 μM) dose‐dependently increases tau phosphorylation at AD‐specific phosphoepitopes in differentiated N2a/p35 cells compared with controls, an effect that is blocked by antisense oligonucleotides against p35. sAβ1–42‐induced tau phosphorylation is concomitant with an increase in both p25 to p35 ratio and Cdk5 activity (but not protein levels). Additionally, blockade of L‐type calcium channels or inhibition of calpain completely abolishes this effect. Taken together, these data indicate that sAβ is a potent activator of the p25/Cdk5 pathway, resulting in promotion of AD‐like tau phosphorylation in vitro.


Journal of Cellular and Molecular Medicine | 2009

Flavonoid-mediated presenilin-1 phosphorylation reduces Alzheimer's disease β-amyloid production

Kavon Rezai-Zadeh; R. Douglas Shytle; Yun Bai; Jun Tian; Huayan Hou; Takashi Mori; Jin Zeng; Demian Obregon; Terrence Town; Jun Tan

Glycogen synthase kinase 3 (GSK‐3) dysregulation is implicated in the two Alzheimers disease (AD) pathological hallmarks: β‐amyloid plaques and neurofibrillary tangles. GSK‐3 inhibitors may abrogate AD pathology by inhibiting amyloidogenic γ‐secretase cleavage of amyloid precursor protein (APP). Here, we report that the citrus bioflavonoid luteolin reduces amyloid‐β (Aβ) peptide generation in both human ‘Swedish’ mutant APP transgene‐bearing neuron‐like cells and primary neurons. We also find that luteolin induces changes consistent with GSK‐3 inhibition that (i) decrease amyloidogenic γ‐secretase APP processing, and (ii) promote presenilin‐1 (PS1) carboxyl‐terminal fragment (CTF) phosphorylation. Importantly, we find GSK‐3α activity is essential for both PS1 CTF phosphorylation and PS1‐APP interaction. As validation of these findings in vivo, we find that luteolin, when applied to the Tg2576 mouse model of AD, decreases soluble Aβ levels, reduces GSK‐3 activity, and disrupts PS1‐APP association. In addition, we find that Tg2576 mice treated with diosmin, a glycoside of a flavonoid structurally similar to luteolin, display significantly reduced Aβ pathology. We suggest that GSK‐3 inhibition is a viable therapeutic approach for AD by impacting PS1 phosphorylation‐dependent regulation of amyloidogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Transcutaneous β-amyloid immunization reduces cerebral β-amyloid deposits without T cell infiltration and microhemorrhage

William V. Nikolic; Yun Bai; Demian Obregon; Huayan Hou; Takashi Mori; Jin Zeng; Jared Ehrhart; R. Douglas Shytle; Brian Giunta; Dave Morgan; Terrence Town; Jun Tan

Alzheimers disease (AD) immunotherapy accomplished by vaccination with β-amyloid (Aβ) peptide has proved efficacious in AD mouse models. However, “active” Aβ vaccination strategies for the treatment of cerebral amyloidosis without concurrent induction of detrimental side effects are lacking. We have developed a transcutaneous (t.c.) Aβ vaccination approach and evaluated efficacy and monitored for deleterious side effects, including meningoencephalitis and microhemorrhage, in WT mice and a transgenic mouse model of AD. We demonstrate that t.c. immunization of WT mice with aggregated Aβ1–42 plus the adjuvant cholera toxin (CT) results in high-titer Aβ antibodies (mainly of the Ig G1 class) and Aβ1–42-specific splenocyte immune responses. Confocal microscopy of the t.c. immunization site revealed Langerhans cells in areas of the skin containing the Aβ1–42 immunogen, suggesting that these unique innate immune cells participate in Aβ1–42 antigen processing. To evaluate the efficacy of t.c. immunization in reducing cerebral amyloidosis, transgenic PSAPP (APPsw, PSEN1dE9) mice were immunized with aggregated Aβ1–42 peptide plus CT. Similar to WT mice, PSAPP mice showed high Aβ antibody titers. Most importantly, t.c. immunization with Aβ1–42 plus CT resulted in significant decreases in cerebral Aβ1–40,42 levels coincident with increased circulating levels of Aβ1–40,42, suggesting brain-to-blood efflux of Aβ. Reduction in cerebral amyloidosis was not associated with deleterious side effects, including brain T cell infiltration or cerebral microhemorrhage. Together, these data suggest that t.c. immunization constitutes an effective and potentially safe treatment strategy for AD.


Nature Communications | 2012

Soluble amyloid precursor protein-α modulates β-secretase activity and amyloid-β generation

Demian Obregon; Huayan Hou; Juan Deng; Brian Giunta; Jun Tian; Donna Darlington; Shahaduzzaman; Yuyuan Zhu; Takashi Mori; Mark P. Mattson; Jun Tan

In sporadic age-related forms of Alzheimers disease (AD), it is unclear why amyloid-β (Aβ) peptides accumulate. Here we show that soluble amyloid precursor protein-α (sAPP-α) decreases Aβ generation by directly associating with β-site APP-converting enzyme (BACE)1, thereby modulating APP processing. Whereas specifically targeting sAPP-α using antibodies enhances Aβ production; in transgenic mice with AD-like pathology, sAPP-α overexpression decreases β-amyloid plaques and soluble Aβ. In support, immunoneutralization of sAPP-α increases APP amyloidogenic processing in these mice. Given our current findings, and because a number of risk factors for sporadic AD serve to lower levels of sAPP-α in brains of AD patients, inadequate sAPP-α levels may be sufficient to polarize APP processing towards the amyloidogenic, Aβ-producing route. Therefore, restoration of sAPP-α or enhancement of its association with BACE may be viable strategies to ameliorate imbalances in APP processing that can lead to AD pathogenesis.


Journal of Neuroinflammation | 2012

The immunology of traumatic brain injury: a prime target for Alzheimer’s disease prevention

Brian Giunta; Demian Obregon; Renuka Velisetty; Paul R. Sanberg; Cesar V. Borlongan; Jun Tan

A global health problem, traumatic brain injury (TBI) is especially prevalent in the current era of ongoing world military conflicts. Its pathological hallmark is one or more primary injury foci, followed by a spread to initially normal brain areas via cascades of inflammatory cytokines and chemokines resulting in an amplification of the original tissue injury by microglia and other central nervous system immune cells. In some cases this may predispose individuals to later development of Alzheimer’s disease (AD). The inflammatory-based progression of TBI has been shown to be active in humans for up to 17 years post TBI. Unfortunately, all neuroprotective drug trials have failed, and specific treatments remain less than efficacious. These poor results might be explained by too much of a scientific focus on neurons without addressing the functions of microglia in the brain, which are at the center of proinflammatory cytokine generation. To address this issue, we provide a survey of the TBI-related brain immunological mechanisms that may promote progression to AD. We discuss these immune and microglia-based inflammatory mechanisms involved in the progression of post-trauma brain damage to AD. Flavonoid-based strategies to oppose the antigen-presenting cell-like inflammatory phenotype of microglia will also be reviewed. The goal is to provide a rationale for investigations of inflammatory response following TBI which may represent a pathological link to AD. In the end, a better understanding of neuroinflammation could open therapeutic avenues for abrogation of secondary cell death and behavioral symptoms that may mediate the progression of TBI to later AD.

Collaboration


Dive into the Demian Obregon's collaboration.

Top Co-Authors

Avatar

Jun Tan

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Brian Giunta

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Huayan Hou

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Takashi Mori

Saitama Medical University

View shared research outputs
Top Co-Authors

Avatar

Terrence Town

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Jared Ehrhart

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jin Zeng

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Jun Tian

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

William V. Nikolic

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge