Network


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

Hotspot


Dive into the research topics where Donna M. Porter is active.

Publication


Featured researches published by Donna M. Porter.


Neuropharmacology | 2002

Anabolic androgenic steroids induce age-, sex-, and dose-dependent changes in GABAA receptor subunit mRNAs in the mouse forebrain

K.L McIntyre; Donna M. Porter; Leslie P. Henderson

Chronic exposure to anabolic androgenic steroids (AAS) has deleterious effects on reproductive health in both human and animal subjects. Neurotransmission mediated by the gamma-aminobutyric acid type A (GABA(A)) receptor in the medial amygdala (MeA), the medial preoptic area (mPOA), and the ventromedial nucleus (VMN) of the hypothalamus plays a critical role in mediating sexual behaviors. Here we used semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR) to examine levels of alpha(1), alpha(2), alpha(5), gamma(1), gamma(2), and epsilon subunit mRNAs in these three regions of the brain. Our results demonstrate that chronic exposure to either a high or a moderate dose of the AAS, 17alpha-methyltestosterone (17alpha-MeT), significantly decreased the levels of specific alpha and gamma subunit mRNAs in a manner that depended on the dose of AAS and age and sex of the animals. Specifically, the moderate dose of AAS elicited significant changes only in pubertal females and the majority of changes observed in pubertal animals with the high dose also occurred in females. In contrast, the moderate dose of AAS induced no significant changes in adult mice of either sex, while the high dose had effects in both males and females. In addition to determining the effects of chronic AAS treatment, a developmental analysis of drug-naïve animals demonstrated that GABA(A) receptor subunit mRNA levels in these regions of the forebrain undergo significant changes as animals proceed through puberty. These data demonstrate that the effects of AAS exposure on GABA(A) receptor expression are superimposed upon dynamic developmental changes that accompany the transition from puberty to adulthood.


Human Molecular Genetics | 2012

A novel mouse model of Niemann–Pick type C disease carrying a D1005G-Npc1 mutation comparable to commonly observed human mutations

Robert A. Maue; Robert W. Burgess; Bing Wang; Christine M. Wooley; Kevin L. Seburn; Marie T. Vanier; Maximillian A. Rogers; Catherine C. Y. Chang; Ta-Yuan Chang; Brent T. Harris; David J. Graber; Carlos A. A. Penatti; Donna M. Porter; Benjamin S. Szwergold; Leslie P. Henderson; John W. Totenhagen; Theodore P. Trouard; Ivan A. Borbon; Robert P. Erickson

We have identified a point mutation in Npc1 that creates a novel mouse model (Npc1(nmf164)) of Niemann-Pick type C1 (NPC) disease: a single nucleotide change (A to G at cDNA bp 3163) that results in an aspartate to glycine change at position 1005 (D1005G). This change is in the cysteine-rich luminal loop of the NPC1 protein and is highly similar to commonly occurring human mutations. Genetic and molecular biological analyses, including sequencing the Npc1(spm) allele and identifying a truncating mutation, confirm that the mutation in Npc1(nmf164) mice is distinct from those in other existing mouse models of NPC disease (Npc1(nih), Npc1(spm)). Analyses of lifespan, body and spleen weight, gait and other motor activities, as well as acoustic startle responses all reveal a more slowly developing phenotype in Npc1(nmf164) mutant mice than in mice with the null mutations (Npc1(nih), Npc1(spm)). Although Npc1 mRNA levels appear relatively normal, Npc1(nmf164) brain and liver display dramatic reductions in Npc1 protein, as well as abnormal cholesterol metabolism and altered glycolipid expression. Furthermore, histological analyses of liver, spleen, hippocampus, cortex and cerebellum reveal abnormal cholesterol accumulation, glial activation and Purkinje cell loss at a slower rate than in the Npc1(nih) mouse model. Magnetic resonance imaging studies also reveal significantly less demyelination/dysmyelination than in the null alleles. Thus, although prior mouse models may correspond to the severe infantile onset forms of NPC disease, Npc1(nmf164) mice offer many advantages as a model for the late-onset, more slowly progressing forms of NPC disease that comprise the large majority of human cases.


The Journal of Neuroscience | 2010

Altered GABAA Receptor-Mediated Synaptic Transmission Disrupts the Firing of Gonadotropin-Releasing Hormone Neurons in Male Mice under Conditions That Mimic Steroid Abuse

Carlos A. A. Penatti; Matthew C. Davis; Donna M. Porter; Leslie P. Henderson

Gonadotropin-releasing hormone (GnRH) neurons are the central regulators of reproduction. GABAergic transmission plays a critical role in pubertal activation of pulsatile GnRH secretion. Self-administration of excessive doses of anabolic androgenic steroids (AAS) disrupts reproductive function and may have critical repercussions for pubertal onset in adolescent users. Here, we demonstrate that chronic treatment of adolescent male mice with the AAS 17α-methyltestosterone significantly decreased action potential frequency in GnRH neurons, reduced the serum gonadotropin levels, and decreased testes mass. AAS treatment did not induce significant changes in GABAA receptor subunit mRNA levels or alter the amplitude or decay kinetics of GABAA receptor-mediated spontaneous postsynaptic currents (sPSCs) or tonic currents in GnRH neurons. However, AAS treatment significantly increased action potential frequency in neighboring medial preoptic area (mPOA) neurons and GABAA receptor-mediated sPSC frequency in GnRH neurons. In addition, physical isolation of the more lateral aspects of the mPOA from the medially localized GnRH neurons abrogated the AAS-induced increase in GABAA receptor-mediated sPSC frequency and the decrease in action potential firing in the GnRH cells. Our results indicate that AAS act predominantly on steroid-sensitive presynaptic neurons within the mPOA to impart significant increases in GABAA receptor-mediated inhibitory tone onto downstream GnRH neurons, resulting in diminished activity of these pivotal mediators of reproductive function. These AAS-induced changes in central GABAergic circuits of the forebrain may significantly contribute to the disruptive actions of these drugs on pubertal maturation and the development of reproductive competence in male steroid abusers.


Brain Research | 2006

Sex-and age-specific effects of anabolic androgenic steroids on reproductive behaviors and on GABAergic transmission in neuroendocrine control regions

Ann S. Clark; Beth A. Costine; Brian L. Jones; Megan C. Kelton-Rehkopf; Sarah H. Meerts; Lora L. Nutbrown-Greene; Carlos A. A. Penatti; Donna M. Porter; Paul Yang; Leslie P. Henderson

Illicit use of anabolic androgenic steroids (AAS) has become a prevalent health concern not only among male professional athletes, but, disturbingly, among a growing number of women and adolescent girls. Despite the increasing use of AAS among women and adolescents, few studies have focused on the effects of these steroids in females, and female adolescent subjects are particularly underrepresented. Among the hallmarks of AAS abuse are changes in reproductive behaviors. Here, we discuss work from our laboratories on the actions of AAS on the onset of puberty and sexual behaviors in female rodents, AAS interactions and sex- and age-specific effects of these steroids on neural transmission mediated by gamma-aminobutyric acid receptors within forebrain neuroendocrine control regions that may underlie AAS-induced changes in these behaviors.


The Journal of Neuroscience | 2009

Chronic Exposure to Anabolic Androgenic Steroids Alters Neuronal Function in the Mammalian Forebrain via Androgen Receptor- and Estrogen Receptor-Mediated Mechanisms

Carlos A. A. Penatti; Donna M. Porter; Leslie P. Henderson

Anabolic androgenic steroids (AAS) can promote detrimental effects on social behaviors for which GABA type A (GABAA) receptor-mediated circuits in the forebrain play a critical role. While all AAS bind to androgen receptors (AR), they may also be aromatized to estrogens and thus potentially impart effects via estrogen receptors (ER). Chronic exposure of wild-type male mice to a combination of chemically distinct AAS increased action potential (AP) frequency, selective GABAA receptor subunit mRNAs, and GABAergic synaptic current decay in the medial preoptic area (mPOA). Experiments performed with pharmacological agents and in AR-deficient Tfm mutant mice suggest that the AAS-dependent enhancement of GABAergic transmission in wild-type mice is AR-mediated. In AR-deficient mice, the AAS elicited dramatically different effects, decreasing AP frequency, spontaneous IPSC amplitude and frequency and the expression of selective GABAA receptor subunit mRNAs. Surprisingly, in the absence of AR signaling, the data indicate that the AAS do not act as ER agonists, but rather suggest a novel in vivo action in which the AAS inhibit aromatase and impair endogenous ER signaling. These results show that the AAS have the capacity to alter neuronal function in the forebrain via multiple steroid signaling mechanisms and suggest that effects of these steroids in the brain will depend not only on the balance of AR- versus ER-mediated regulation for different target genes, but also on the ability of these drugs to alter steroid metabolism and thus the endogenous steroid milieu.


Clinical Endocrinology | 2005

Multiple endocrine neoplasia 2A due to a unique C609S RET mutation presents with pheochromocytoma and reduced penetrance of medullary thyroid carcinoma

William B. Kinlaw; Sarah M. Scott; Robert A. Maue; Vincent A. Memoli; Robert D. Harris; Gilbert H. Daniels; Donna M. Porter; Dorothy R. Belloni; Edward T. Spooner; Manfred M. Ernesti; Walter W. Noll

Objective   We have identified a large kindred with multiple endocrine neoplasia 2A (MEN 2A) due to a mutation at RET codon 609 that results in a cysteine to serine substitution, a mutation previously identified in only one case in the literature. We characterized the clinical phenotype of the kindred and the biochemical mechanism of this new mutation.


Psychoneuroendocrinology | 2010

Chronic anabolic androgenic steroid exposure alters corticotropin releasing factor expression and anxiety-like behaviors in the female mouse

Beth A. Costine; Joseph G. Oberlander; Matthew C. Davis; Carlos A. A. Penatti; Donna M. Porter; Robert N. Leaton; Leslie P. Henderson

In the past several decades, the therapeutic use of anabolic androgenic steroids (AAS) has been overshadowed by illicit use of these drugs by elite athletes and a growing number of adolescents to enhance performance and body image. As with adults, AAS use by adolescents is associated with a range of behavioral effects, including increased anxiety and altered responses to stress. It has been suggested that adolescents, especially adolescent females, may be particularly susceptible to the effects of these steroids, but few experiments in animal models have been performed to test this assertion. Here we show that chronic exposure of adolescent female mice to a mixture of three commonly abused AAS (testosterone cypionate, nandrolone decanoate and methandrostenolone; 7.5 mg/kg/day for 5 days) significantly enhanced anxiety-like behavior as assessed by the acoustic startle response (ASR), but did not augment the fear-potentiated startle response (FPS) or alter sensorimotor gating as assessed by prepulse inhibition of the acoustic startle response (PPI). AAS treatment also significantly increased the levels of corticotropin releasing factor (CRF) mRNA and somal-associated CRF immunoreactivity in the central nucleus of the amygdala (CeA), as well as neuropil-associated immunoreactivity in the dorsal aspect of the anterolateral division of the bed nucleus of the stria terminalis (dBnST). AAS treatment did not alter CRF receptor 1 or 2 mRNA in either the CeA or the dBnST; CRF immunoreactivity in the ventral BnST, the paraventricular nucleus (PVN) or the median eminence (ME); or peripheral levels of corticosterone. These results suggest that chronic AAS treatment of adolescent female mice may enhance generalized anxiety, but not sensorimotor gating or learned fear, via a mechanism that involves increased CRF-mediated signaling from CeA neurons projecting to the dBnST.


Neuroscience | 2005

Sex-specific effects of chronic anabolic androgenic steroid treatment on GABAA receptor expression and function in adolescent mice

Carlos A. A. Penatti; Donna M. Porter; Brian L. Jones; Leslie P. Henderson

Anabolic androgenic steroids are synthetic derivatives of testosterone designed for therapeutic uses, but now taken as drugs of abuse. Potential health risks associated with anabolic androgenic steroid abuse are believed to be higher in adolescents than in adults, but few studies have tested anabolic androgenic steroid effects in adolescent subjects or determined if effects of these steroids differ between females and males. We have studied GABA(A) receptor expression and function in the medial preoptic nucleus of mice chronically treated during adolescence with the anabolic androgenic steroid, 17alpha-methyltestosterone. Three-week treatment did not elicit significant differences the expression of alpha1, alpha2 or alpha5 subunit mRNAs in animals of either sex, although there was a trend toward decreases in all three subunit mRNAs in female mice, which was augmented and attained significance for the alpha2 subunit mRNA in females treated for six weeks. Immunocytochemical analysis revealed that treatment with 17alpha-methyltestosterone for 6 weeks also elicited a significant decrease in the number of alpha2-immunopositive neurons in female subjects. To test if anabolic androgenic steroid treatment also promoted changes in GABA(A) receptor function, spontaneous inhibitory synaptic currents were analyzed in adolescent animals treated for 3-4 weeks. This treatment regimen promoted a significant decrease in spontaneous inhibitory synaptic current frequency in female, but not male mice. Finally, anabolic androgenic steroid treatment was found to have no effect on the numbers of interneurons within the medial preoptic nucleus, as assessed by immunoreactivity for calcium binding proteins, suggesting that the decrease in the frequency of spontaneous inhibitory synaptic currents in female mice does not arise from an anabolic androgenic steroid-induced loss of interneurons. Taken together, our results indicate that chronic exposure to 17alpha-methyltestosterone elicits significant changes in GABAergic transmission in the medial preoptic nucleus of female, but not male, mice effectively enhancing the sexually dimorphic nature of GABAergic transmission in a forebrain region crucial for the expression of aggression and sexual behaviors.


Hormones and Behavior | 2003

The display of sexual behaviors by female rats administered ICI 182,780.

Ann S. Clark; Fay A. Guarraci; Alison B. Megroz; Donna M. Porter; Leslie P. Henderson

ICI 182,780 (ICI) is a pure antiestrogen that when administered systemically does not cross the blood-brain barrier, thus its actions are limited to the periphery. Four experiments were conducted to test the effects of ICI on the display of sexual behaviors in ovariectomized rats. Experiment 1 examined the effects of three doses of ICI (250, 500, and 750 micro g/rat) on sexual receptivity and paced mating behavior in rats primed with estradiol benzoate (EB) in combination with progesterone (P). Experiments 2 and 3 compared the display of sexual behaviors in rats primed with EB+P or EB alone and administered either 250 micro g ICI (Experiment 2) or 500 micro g ICI (Experiment 3). Experiment 4 tested the effects of ICI (250 and 500 micro g) on the expression of estrogen-induced progestin receptors in the uterus. ICI did not affect the display of sexual receptivity in any experiment. In rats primed with EB+P, paced mating behavior was altered by the 500 and 750 micro g, but not the 250 micro g, doses of ICI. The lowest (250 micro g) dose of ICI did alter paced mating behavior in rats primed with EB alone. The effects of ICI on paced mating behavior were manifested by a substantial lengthening of contact-return latencies following intromissions and ejaculations. The percentage of exits were not affected by ICI. Estrogen stimulation of uterine weight and induction of uterine progestin receptors was suppressed by ICI (250 and 500 micro g). ICI effects on paced mating behavior in hormone-primed female rats are likely to reflect antiestrogenic actions in the periphery, including interference with the estrogen induction of progestin receptors.


Neuroscience | 2009

Effects of chronic exposure to an anabolic androgenic steroid cocktail on α5-receptor-mediated GABAergic transmission and neural signaling in the forebrain of female mice

Carlos A. A. Penatti; Beth A. Costine; Donna M. Porter; Leslie P. Henderson

Anabolic androgenic steroids (AAS) are synthetic derivatives of testosterone that are illicitly self-administered for enhancement of performance and body image, but which also have significant effects on the brain and on behavior. While the stereotypical AAS user is an adult male, AAS abuse in women is rapidly increasing, yet few studies have examined AAS effects in female subjects. We have assessed the effects in female mice of a combination of commonly abused AAS on neuronal activity and neurotransmission mediated by GABA type A (GABA(A)) receptors in the medial preoptic nucleus (MPN); a nexus in the circuits of the hypothalamus and forebrain that are critical for the expression of social behaviors known to be altered in AAS abuse. Our data indicate that chronic exposure to AAS resulted in androgen receptor (AR)-dependent upregulation of alpha(5), beta(3) and delta subunit mRNAs. Acute application of the alpha(5) subunit-selective inverse agonist, L-655,708 (L6), indicated that a significant fraction of the synaptic current is carried by alpha(5)-containing receptors and that AAS treatment may enhance expression of alpha(5)-containing receptors contributing to synaptic, but not tonic, currents in the MPN. AAS treatment also resulted in a significant decrease in action potential frequency in MPN neurons that was also correlated with an increased sensitivity to L-655,708. Our data demonstrate that chronic exposure to multiple AAS elicits significant changes in GABAergic transmission and neuronal activity that are likely to reflect changes in the expression of alpha(5)-containing synaptic receptors within the MPN.

Collaboration


Dive into the Donna M. Porter's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge