Network


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

Hotspot


Dive into the research topics where Delinda A. Johnson is active.

Publication


Featured researches published by Delinda A. Johnson.


The Journal of Neuroscience | 2004

Nuclear Factor E2-Related Factor 2-Dependent Antioxidant Response Element Activation by tert-Butylhydroquinone and Sulforaphane Occurring Preferentially in Astrocytes Conditions Neurons against Oxidative Insult

Andrew D. Kraft; Delinda A. Johnson; Jeffrey A. Johnson

Binding of the transcription factor nuclear factor E2-related factor 2 (Nrf2) to the antioxidant response element (ARE) in neural cells results in the induction of a battery of genes that can coordinate a protective response against a variety of oxidative stressors. In this study, tert-butylhydroquinone (tBHQ) and sulforaphane were used as activators of this pathway. Consistent with previous studies, treatment of primary cortical cultures from ARE reporter mice revealed selective promoter activity in astrocytes. This activation protected neurons from hydrogen peroxide and nonexcitotoxic glutamate toxicity. tBHQ treatment of cultures from Nrf2 knock-out animals resulted in neither ARE activation nor neuroprotection. By reintroducing Nrf2 via infection with a replication-deficient adenovirus (ad), both the genetic response and neuroprotection were rescued. Conversely, infection with adenovirus encoding dominant-negative (DN) Nrf2 (ad-DN-Nrf2) or pretreatment with the selective phosphatidylinositol-3 kinase inhibitor LY294002 inhibited the tBHQ-mediated promoter response and corresponding neuroprotection. Interestingly, the adenoviral infection showed a high selectivity for astrocytes over neurons. In an attempt to reveal some of the cell type-specific changes resulting from ARE activation, cultures were infected with adenovirus encoding green fluorescent protein (GFP) (ad-GFP) or ad-DN-Nrf2 (containing GFP) before tBHQ treatment. A glia-enriched population of GFP-infected cells was then isolated from a population of uninfected neurons using cell-sorting technology. Microarray analysis was used to evaluate potential glial versus neuron-specific contributions to the neuroprotective effects of ARE activation and Nrf2 dependence. Strikingly, the change in neuronal gene expression after tBHQ treatment was dependent on Nrf2 activity in the astrocytes. This suggests that Nrf2-dependent genetic changes alter neuron–glia interactions resulting in neuroprotection.


The FASEB Journal | 2005

Nrf2, a multi-organ protector?

Jong-Min Lee; Delinda A. Johnson; Thor D. Stein; Andrew D. Kraft; Marcus J. Calkins; Rebekah J. Jakel; Jeffrey A. Johnson

NF‐E2‐related factor 2 (Nrf2) is a basic leucine zipper transcription factor that binds to the promoter sequence “antioxidant responsive element (ARE)” leading to coordinated up‐regulation of ARE‐driven detoxification and antioxidant genes. Since the expression of a wide array of antioxidant and detoxification genes are positively regulated by the ARE sequence, Nrf2 may serve as a master regulator of the ARE‐driven cellular defense system against oxidative stress. In support of this, numerous studies have shown that Nrf2 protects many cell types and organ systems from a broad spectrum of toxic insults and disease pathogenesis. This Nrf2‐conferred, multi‐organ protection phenomenon raises an interesting question about how a single protein can protect many different organs from various toxic insults. A possible molecular mechanism explaining this phenomenon is that Nrf2 protects many different cell types by coordinately up‐regulating classic ARE‐driven genes as well as cell type‐specific target genes that are required for the defense system of each cell type in its unique environment. This hypothesis is supported by microarray data indicating the protective role of Nrf2 is conveyed through both known ARE‐driven genes and novel cell type‐specific genes. The widespread nature of Nrf2 may have an important therapeutic potential, allowing prevention of carcinogenesis and neurodegenerative diseases. Lee, J.‐M., Li, J., Johnson, D. A., Stein, T. D., Kraft, A. D., Calkins, M. J., Jakel, R. J., Johnson, J. A. Nrf2, a multi‐organ protector? FASEB J. 19, 1061–1066 (2005)


Proceedings of the National Academy of Sciences of the United States of America | 2009

Nrf2-mediated neuroprotection in the MPTP mouse model of Parkinson's disease: Critical role for the astrocyte

Pei Chun Chen; Marcelo R. Vargas; Amar K. Pani; Richard J. Smeyne; Delinda A. Johnson; Yuet Wai Kan; Jeffrey A. Johnson

Oxidative stress has been implicated in the etiology of Parkinsons disease (PD) and in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) animal model of PD. It is known that under conditions of oxidative stress, the transcription factor NF-E2-related factor (Nrf2) binds to antioxidant response element (ARE) to induce antioxidant and phase II detoxification enzymes. To investigate the role of Nrf2 in the process of MPTP-induced toxicity, mice expressing the human placental alkaline phosphatase (hPAP) gene driven by a promoter containing a core ARE sequence (ARE-hPAP) were used. ARE-hPAP mice were injected (30 mg/kg) once per day for 5 days and killed 7 days after the last MPTP injection. In response to this design, ARE-dependent gene expression was decreased in striatum whereas it was increased in substantia nigra. The same MPTP protocol was applied in Nrf2+/+ and Nrf2−/− mice; Nrf2 deficiency increases MPTP sensitivity. Furthermore, we evaluated the potential for astrocytic Nrf2 overexpression to protect from MPTP toxicity. Transgenic mice with Nrf2 under control of the astrocyte-specific promoter for the glial fribillary acidic protein (GFAP-Nrf2) on both a Nrf2+/+ and Nrf2−/− background were administered MPTP. In the latter case, only the astrocytes expressed Nrf2. Independent of background, MPTP-mediated toxicity was abolished in GFAP-Nrf2 mice. These striking results indicate that Nrf2 expression restricted to astrocytes is sufficient to protect against MPTP and astrocytic modulation of the Nrf2-ARE pathway is a promising target for therapeutics aimed at reducing or preventing neuronal death in PD.


Annals of the New York Academy of Sciences | 2008

The Nrf2–ARE Pathway

Jeffrey A. Johnson; Delinda A. Johnson; Andrew D. Kraft; Marcus J. Calkins; Rebekah J. Jakel; Marcelo R. Vargas; Pei Chun Chen

Transcriptional activation of protective genes is mediated by a cis‐acting element called the antioxidant responsive element (ARE). The transcription factor Nrf2 (NF–E2‐related factor 2) binds to the ARE. Activation of this pathway protects cells from oxidative stress‐induced cell death. Increased oxidative stress is associated with neuronal cell death during the pathogenesis of multiple chronic neurodegenerative diseases, including Alzheimers disease, Parkinsons disease, Huntingtons disease, and amyotrophic lateral sclerosis. We hypothesize that Nrf2–ARE activation is a novel neuroprotective pathway that confers resistance to a variety of oxidative, stress‐related, neurodegenerative insults. In recent studies, primary neuronal cultures treated with chemical activators of the Nrf2–ARE pathway displayed significantly greater resistance to oxidative stress‐induced neurotoxicity. Similar cultures generated from ARE–hPAP reporter mice demonstrated selective activation of the Nrf2–ARE pathway in astrocytes, suggesting that Nrf2 activation in astrocytes somehow confers resistance to naive neurons. Further, in chemical models of neurodegeneration, Nrf2 knockout mice are significantly more sensitive to mitochondrial complex I and II inhibitors. Combining these observations with the results implying that the astrocyte is central to Nrf2–ARE‐mediated neuroprotection, we transplanted Nrf2‐overexpressing astrocytes into the mouse striatum prior to lesioning with malonate. This procedure led to dramatic protection against malonate‐induced neurotoxicity. Translating this to other chemical and genetic models of neurodegeneration will be discussed.


The Journal of Neuroscience | 2008

Nrf2 Activation in Astrocytes Protects against Neurodegeneration in Mouse Models of Familial Amyotrophic Lateral Sclerosis

Marcelo R. Vargas; Delinda A. Johnson; Daniel W. Sirkis; Albee Messing; Jeffrey A. Johnson

Activation of the transcription factor Nrf2 in astrocytes coordinates the upregulation of antioxidant defenses and confers protection to neighboring neurons. Dominant mutations in Cu/Zn-superoxide dismutase (SOD1) cause familial forms of amyotrophic lateral sclerosis (ALS), a fatal disorder characterized by the progressive loss of motor neurons. Non-neuronal cells, including astrocytes, shape motor neuron survival in ALS and are a potential target to prevent motor neuron degeneration. The protective effect of Nrf2 activation in astrocytes has never been examined in a chronic model of neurodegeneration. We generated transgenic mice over-expressing Nrf2 selectively in astrocytes using the glial fibrillary acidic protein (GFAP) promoter. The toxicity of astrocytes expressing ALS-linked mutant hSOD1 to cocultured motor neurons was reversed by Nrf2 over-expression. Motor neuron protection depended on increased glutathione secretion from astrocytes. This protective effect was also observed by crossing the GFAP-Nrf2 mice with two ALS-mouse models. Over-expression of Nrf2 in astrocytes significantly delayed onset and extended survival. These findings demonstrate that Nrf2 activation in astrocytes is a viable therapeutic target to prevent chronic neurodegeneration.


Molecular Cancer Therapeutics | 2007

Pharmacodynamic characterization of chemopreventive triterpenoids as exceptionally potent inducers of Nrf2-regulated genes

Melinda S. Yates; Masafumi Tauchi; Fumiki Katsuoka; Kathleen C. Flanders; Karen T. Liby; Tadashi Honda; Gordon W. Gribble; Delinda A. Johnson; Jeffrey A. Johnson; Neal C. Burton; Tomás R. Guilarte; Masayuki Yamamoto; Michael B. Sporn; Thomas W. Kensler

Synthetic triterpenoids have been developed, which are potent inducers of cytoprotective enzymes and inhibitors of inflammation, greatly improving on the weak activity of naturally occurring triterpenoids. An imidazolide triterpenoid derivative, 1-[2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oyl]imidazole (CDDO-Im or TP235), has been previously shown to potently protect against hepatic tumorigenesis, acting in part by inducing cytoprotective genes through Keap1-Nrf2-antioxidant response element (ARE) signaling. In these studies, the pharmacodynamic activity of CDDO-Im is characterized in two distinct lines of ARE reporter mice and by measuring increases in Nqo1 transcript levels as a marker of cytoprotective gene induction. Oral administration of CDDO-Im induces ARE-regulated cytoprotective genes in many tissues in the mouse, including liver, lung, kidney, intestines, brain, heart, thymus, and salivary gland. CDDO-Im induces Nqo1 RNA transcripts in some organs at doses as low as 0.3 μmol/kg body weight (orally). A structure activity evaluation of 15 additional triterpenoids (a) confirmed the importance of Michael acceptor groups on both the A and C rings, (b) showed the requirement for a nitrile group at C-2 of the A ring, and (c) indicated that substituents at C-17 dramatically affected pharmacodynamic action in vivo. In addition to CDDO-Im, other triterpenoids, particularly the methyl ester CDDO-Me (TP155) and the dinitrile TP225, are extremely potent inducers of cytoprotective genes in mouse liver, lung, small intestine mucosa, and cerebral cortex. This pharmacodynamic characterization highlights the chemopreventive promise of several synthetic triterpenoids in multiple target organs. [Mol Cancer Ther 2007;6(1):154–62]


Journal of Neurochemistry | 2002

Activation of the antioxidant response element in primary cortical neuronal cultures derived from transgenic reporter mice

Delinda A. Johnson; G. K. Andrews; W. Xu; Jeffrey A. Johnson

Many phase II protective genes contain a cis‐acting enhancer region known as the antioxidant response element (ARE). Increased expression of these genes contributes to the protection of cells from oxidative stress. Transgenic reporter mice were created that carry in their genome the core ARE coupled to the human placental alkaline phosphatase (hPAP) reporter gene. Primary cortical cultures derived from these mice were treated with tBHQ resulting in a dose‐dependent increase in hPAP activity. Histochemical staining for hPAP activity was observed in both glia and neurons from tBHQ‐treated cultures. The tBHQ‐mediated increase in hPAP was not affected by the antioxidant glutathione monoethyl ester (GSHEE), whereas the increase in hPAP following DEM treatment was completely blocked by GSHEE. Pre‐treatment of cultures with the PI3‐kinase inhibitor LY 294002 demonstrated a dose‐dependent decrease in tBHQ‐induced hPAP activity. In addition, the tBHQ‐mediated expression of ARE‐driven genes in primary cortical cultures was blocked by LY 294002. Interestingly, basal expression of Nrf2 was also inhibited by LY 294002. We theorize that increased levels of genes controlled by the ARE are important for cellular protection against oxidative stress. These ARE‐hPAP transgenic mice will be an important in vivo model for testing our hypothesis.


Journal of Neurochemistry | 2010

Plumbagin, a novel Nrf2/ARE activator, protects against cerebral ischemia

Tae Gen Son; Simonetta Camandola; Thiruma V. Arumugam; Roy G. Cutler; Richard Telljohann; Mohamed R. Mughal; Tyson A. Moore; Weiming Luo; Qian Sheng Yu; Delinda A. Johnson; Jeffrey A. Johnson; Mark P. Mattson

J. Neurochem. (2010) 112, 1316–1326.


Toxicological Sciences | 2010

The Absence of the Pro-antioxidant Transcription Factor Nrf2 Exacerbates Experimental Autoimmune Encephalomyelitis

Delinda A. Johnson; Sara Amirahmadi; Charlotte Ward; Zsuszanna Fabry; Jeffrey A. Johnson

Multiple sclerosis (MS) is an autoimmune disease characterized by peripheral activation of CD4(+) T cells that migrate into the central nervous system (CNS) and mount an autoimmune neuroinflammatory attack on myelin and oligodendrocytes. Secondary to these events, however equally destructive, is the generation of inflammatory-mediated reactive oxygen and nitrogen species generated by persistently activated microglia and astrocytes. Nuclear factor-erythroid 2-related factor 2 (Nrf2) is a basic leucine zipper transcription factor that regulates genetic expression of many protective antioxidant and detoxication enzymes. Here we describe the Nrf2 modulation of innate and adaptive immune responses in an acute autoimmune model of MS, experimental autoimmune encephalomyelitis (EAE). Wild-type (WT) mice and Nrf2 knockout mice were immunized with myelin oligodendrocyte glycoprotein (MOG 35-55) and monitored daily for clinical scores of disease. Disruption of Nrf2 resulted in a more severe clinical course, a more rapid onset, and a greater percentage of mice with the disease. Furthermore, increased immune cell infiltration and glial cell activation in spine was observed. In conjunction, we observed increased inflammatory enzyme (iNOS, phox-47, gp91-phox, and phox-67), cytokine (IFN-gamma, IL1-b, TNF-alpha, and IL-12), and chemokine (BLC and MIG) gene expression levels in the Nrf2-deficient mice compared to the WT mice, supporting the notion that Nrf2 can modulate an autoimmune neuroinflammatory response. Our results show that the absence of Nrf2 exacerbates the development of EAE and thus suggests that activation of Nrf2 may then attenuate pathogenesis of autoimmune diseases such as MS as well as other neurodegenerative diseases that present with neuroinflammation.


Free Radical Biology and Medicine | 2015

Nrf2—a therapeutic target for the treatment of neurodegenerative diseases

Delinda A. Johnson; Jeffrey A. Johnson

The brain is very sensitive to changes in redox status; thus maintaining redox homeostasis in the brain is critical for the prevention of accumulating oxidative damage. Aging is the primary risk factor for developing neurodegenerative diseases. In addition to age, genetic and environmental risk factors have also been associated with disease development. The primary reactive insults associated with the aging process are a result of oxidative stress (OS) and nitrosative stress (NS). Markers of increased oxidative stress, protein and DNA modification, inflammation, and dysfunctional proteostasis have all been implicated in contributing to the progression of neurodegeneration. The ability of the cell to combat OS/NS and maintain a clearance mechanism for misfolded aggregating proteins determines whether or not it will survive. A critical pathway in this regard is the Nrf2 (nuclear factor erythroid 2-related factor 2)- antioxidant response element (ARE) pathway. Nrf2 activation has been shown to mitigate a number of pathologic mechanisms associated with Alzheimers disease, Parkinsons disease, amyotrophic lateral sclerosis, Huntingtons disease, and multiple sclerosis. This review will focus on the role of Nrf2 in these diseases and the potential for Nrf2 activation to attenuate disease progression.

Collaboration


Dive into the Delinda A. Johnson's collaboration.

Top Co-Authors

Avatar

Jeffrey A. Johnson

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Marcus J. Calkins

National Cheng Kung University

View shared research outputs
Top Co-Authors

Avatar

Andrew D. Kraft

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Marcelo R. Vargas

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jong-Min Lee

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Rebekah J. Jakel

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Albee Messing

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

David P. Foley

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Li Gan

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge