Network


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

Hotspot


Dive into the research topics where Andrew M. Allen is active.

Publication


Featured researches published by Andrew M. Allen.


The International Journal of Biochemistry & Cell Biology | 2003

The brain renin-angiotensin system: location and physiological roles

Michael J. McKinley; Anthony L. Albiston; Andrew M. Allen; Michael L. Mathai; Clive N. May; Robin M. McAllen; Brian J. Oldfield; Frederick A.O. Mendelsohn; Siew Yeen Chai

Angiotensinogen, the precursor molecule for angiotensins I, II and III, and the enzymes renin, angiotensin-converting enzyme (ACE), and aminopeptidases A and N may all be synthesised within the brain. Angiotensin (Ang) AT(1), AT(2) and AT(4) receptors are also plentiful in the brain. AT(1) receptors are found in several brain regions, such as the hypothalamic paraventricular and supraoptic nuclei, the lamina terminalis, lateral parabrachial nucleus, ventrolateral medulla and nucleus of the solitary tract (NTS), which are known to have roles in the regulation of the cardiovascular system and/or body fluid and electrolyte balance. Immunohistochemical and neuropharmacological studies suggest that angiotensinergic neural pathways utilise Ang II and/or Ang III as a neurotransmitter or neuromodulator in the aforementioned brain regions. Angiotensinogen is synthesised predominantly in astrocytes, but the processes by which Ang II is generated or incorporated in neurons for utilisation as a neurotransmitter is unknown. Centrally administered AT(1) receptor antagonists or angiotensinogen antisense oligonucleotides inhibit sympathetic activity and reduce arterial blood pressure in certain physiological or pathophysiological conditions, as well as disrupting water drinking and sodium appetite, vasopressin secretion, sodium excretion, renin release and thermoregulation. The AT(4) receptor is identical to insulin-regulated aminopeptidase (IRAP) and plays a role in memory mechanisms. In conclusion, angiotensinergic neural pathways and angiotensin peptides are important in neural function and may have important homeostatic roles, particularly related to cardiovascular function, osmoregulation and thermoregulation.


Brain Research Bulletin | 1998

Angiotensin receptors in the nervous system

Andrew M. Allen; Ingrid Moeller; Trisha A. Jenkins; Jialong Zhuo; G.P. Aldred; Syn Y Chai; Frederick A.O. Mendelsohn

In addition to its traditional role as a circulating hormone, angiotensin is also involved in local functions through the activity of tissue renin-angiotensin systems that occur in many organs, including the brain. In the brain, both systemic and presumptive neurally derived angiotensin and angiotensin metabolites act through specific receptors to modulate many functions. This review examines the distribution of these specific angiotensin receptors and discusses evidence regarding the function of angiotensin peptides in various brain regions. Angiotensin AT1 and AT2 receptors occur in characteristic distributions that are highly correlated with the distribution of angiotensin-like immunoreactivity in nerve terminals. Acting through the AT1 receptor in the brain, angiotensin has effects on fluid and electrolyte homeostasis, neuroendocrine systems, autonomic pathways regulating cardiovascular function and behavior. Angiotensin AT1 receptors are also found in many afferent and efferent components of the peripheral autonomic nervous system. The role of the AT2 receptor in the brain is less well understood, although recent knockout studies point to an involvement with behavioral and cardiovascular functions. In addition to the AT1 and AT2 receptors, receptors for other fragments of angiotensin have been proposed. The AT4 binding site, which binds angiotensin, has a widespread distribution in the brain quite distinct from that of the AT1 and AT2 receptors. It is associated with many cholinergic neuronal groups and also several sensory nuclei, but its function remains to be determined. Our discovery that another brain-derived peptide binds to the AT4 binding site in the brain and may represent the native ligand is discussed. Overall, the distribution of angiotensin receptors in the brain indicate that they play diverse and important physiological roles in the nervous system.


American Journal of Hypertension | 2000

Localization and function of angiotensin AT1 receptors

Andrew M. Allen; Jialong Zhuo; Frederick A.O. Mendelsohn

The distributions of angiotensin AT1 and AT2 receptors have been mapped by in vitro autoradiography throughout most tissues of many mammals, including humans. In addition to confirming that AT1 receptors occur in sites known to be targets for the physiologic actions of angiotensin, such as the adrenal cortex and medulla, renal glomeruli and proximal tubules, vascular and cardiac muscle and brain circumventricular organs, many new sites of action have been demonstrated. In the kidney, AT1 receptors occur in high density in renal medullary interstitial cells. The function of these cells, which span the interstitial space between the tubules and the vasa rectae, remains to be determined. Renal medullary interstitial cells possess receptors for a number of vasoactive hormones in addition to AT1 receptors and this, in concert with their anatomic location, suggests they may be important for the regulation of fluid reabsorption or renal medullary blood flow. In the heart, the highest densities of AT1 receptors occur in association with the conduction system and vagal ganglia. In the central nervous system, high AT1 receptor densities occur in many regions behind the blood-brain barrier, supporting a role for neurally derived angiotensin as a neuromodulator. The physiologic role of angiotensin in many of these brain sites remains to be determined. The AT2 receptor also has a characteristic distribution in several tissues including the adrenal gland, heart, and brain. The role of this receptor in physiology is being elucidated, but it appears to inhibit proliferation and to participate in development. Thus, receptor-binding studies, localizing the distribution of AT1 and AT2 receptors, provide many insights into novel physiologic roles of angiotensin.


Circulation Research | 1991

Localization of angiotensin converting enzyme in rat heart

Hiroshi Yamada; Bruno Fabris; Andrew M. Allen; Bruce Jackson; Colin I. Johnston; A O Mendelsohn

Angiotensin converting enzyme (ACE) was localized in rat heart by quantitative in vitro autoradiography with 125I-351A as the radioligand. The binding association constant (KA) of the radioligand was measured in membrane-rich fractions of atrium, ventricle, and lung by a radioinhibitor binding assay. A single class of high-affinity binding sites was detected in each tissue, and a significant difference was found between KA values for atria and ventricles with a rank order of atria greater than lungs greater than ventricles. For autoradiography, coronal sections (10 micron) of the frozen heart were incubated with 125I-351A and exposed to x-ray film. The autoradiographs were quantitated by computerized image analysis. The highest density of ACE in the heart was found on valve leaflets (aortic, pulmonary, mitral, and tricuspid), which contrasted markedly with very low ACE labeling in the endocardium. The coronary arteries also showed dense labeling of ACE. The right atrium had a moderate density of ACE, which was higher than the left atrium and the ventricles. Both the endothelial and adventitial layers of the aorta and pulmonary artery displayed high densities of ACE, with very low density in the media. ACE was not detected in either the sinoatrial node or atrioventricular node. These results reveal a markedly nonuniform localization of ACE in the rat heart and suggest possible sites for local angiotensin II generation and bradykinin or other peptide metabolism.


Cell Metabolism | 2011

AT1A Angiotensin Receptors in the Renal Proximal Tubule Regulate Blood Pressure

Susan B. Gurley; Anne Riquier-Brison; Jurgen Schnermann; Matthew A. Sparks; Andrew M. Allen; Volker H. Haase; John N. Snouwaert; Thu H. Le; Alicia A. McDonough; Beverley Koller; Thomas M. Coffman

Hypertension affects more than 1.5 billion people worldwide but the precise cause of elevated blood pressure (BP) cannot be determined in most affected individuals. Nonetheless, blockade of the renin-angiotensin system (RAS) lowers BP in the majority of patients with hypertension. Despite its apparent role in hypertension pathogenesis, the key cellular targets of the RAS that control BP have not been clearly identified. Here we demonstrate that RAS actions in the epithelium of the proximal tubule have a critical and nonredundant role in determining the level of BP. Abrogation of AT(1) angiotensin receptor signaling in the proximal tubule alone is sufficient to lower BP, despite intact vascular responses. Elimination of this pathway reduces proximal fluid reabsorption and alters expression of key sodium transporters, modifying pressure-natriuresis and providing substantial protection against hypertension. Thus, effectively targeting epithelial functions of the proximal tubule of the kidney should be a useful therapeutic strategy in hypertension.


The Journal of Physiology | 2009

Amplified respiratory-sympathetic coupling in the spontaneously hypertensive rat: does it contribute to hypertension?

Annabel E. Simms; Julian F. R. Paton; Anthony E. Pickering; Andrew M. Allen

Sympathetic nerve activity (SNA) is elevated in established hypertension. We tested the hypothesis that SNA is elevated in neonate and juvenile spontaneously hypertensive (SH) rats prior to the development of hypertension, and that this may be due to augmented respiratory–sympathetic coupling. Using the working heart–brainstem preparation, perfusion pressure, phrenic nerve activity and thoracic (T8) SNA were recorded in male SH rats and normotensive Wistar–Kyoto (WKY) rats at three ages: neonates (postnatal day 9–16), 3 weeks old and 5 weeks old. Perfusion pressure was higher in SH rats at all ages reflecting higher vascular resistance. The amplitude of respiratory‐related bursts of SNA was greater in SH rats at all ages (P < 0.05). This was reflected in larger Traube–Hering pressure waves in SH rats (1.4 ± 0.8 versus 9.8 ± 1.5 mmHg WKY versus SH rat, 5 weeks old, n= 5 per group, P < 0.01). Recovery from hypocapnic‐induced apnoea and reinstatement of Traube–Hering waves produced a significantly greater increase in perfusion pressure in SH rats (P < 0.05). Differences in respiratory–sympathetic coupling in the SH rat were not secondary to changes in central or peripheral chemoreflex sensitivity, nor were they related to altered arterial baroreflex function. We have shown that increased SNA is already present in SH rats in early postnatal life as revealed by augmented respiratory modulation of SNA. This is reflected in an increased magnitude of Traube–Hering waves resulting in elevated perfusion pressure in the SH rat. We suggest that the amplified respiratory‐related bursts of SNA seen in the neonate and juvenile SH rat may be causal in the development of their hypertension.


Clinical and Experimental Pharmacology and Physiology | 1998

INTERACTION OF CIRCULATING HORMONES WITH THE BRAIN: THE ROLES OF THE SUBFORNICAL ORGAN AND THE ORGANUM VASCULOSUM OF THE LAMINA TERMINALIS

Michael J. McKinley; Andrew M. Allen; Peta Burns; L.M Colvill; Brian J. Oldfield

1. Most circulating peptide hormones are excluded from much of the brain by the blood‐brain barrier. However, they do have access to the circumventricular organs (CVO), which lack the blood‐brain barrier. Three of the CVO, the subfornical organ (SFO), organum vasculosum of the lamina terminalis (OVLT) and area postrema, contain neurons responsive to peptides such as angiotensin II (Angll), atrial natriuretic peptide and relaxin.


Cell | 2014

Leptin mediates the increase in blood pressure associated with obesity

Stephanie E. Simonds; Jack T. Pryor; Eric Ravussin; Frank L. Greenway; Ralph J. DiLeone; Andrew M. Allen; Jaspreet K. Bassi; Joel K. Elmquist; Julia M. Keogh; Elana Henning; Martin G. Myers; Julio Licinio; Russell D. Brown; Pablo J. Enriori; Stephen O’Rahilly; Scott M. Sternson; Kevin L. Grove; David Spanswick; I. Sadaf Farooqi; Michael A. Cowley

Summary Obesity is associated with increased blood pressure (BP), which in turn increases the risk of cardiovascular diseases. We found that the increase in leptin levels seen in diet-induced obesity (DIO) drives an increase in BP in rodents, an effect that was not seen in animals deficient in leptin or leptin receptors (LepR). Furthermore, humans with loss-of-function mutations in leptin and the LepR have low BP despite severe obesity. Leptin’s effects on BP are mediated by neuronal circuits in the dorsomedial hypothalamus (DMH), as blocking leptin with a specific antibody, antagonist, or inhibition of the activity of LepR-expressing neurons in the DMH caused a rapid reduction of BP in DIO mice, independent of changes in weight. Re-expression of LepRs in the DMH of DIO LepR-deficient mice caused an increase in BP. These studies demonstrate that leptin couples changes in weight to changes in BP in mammalian species.


Journal of Hypertension | 1998

Mapping tissue angiotensin-converting enzyme and angiotensin AT1, AT2 and AT4 receptors.

Jialong Zhuo; Ingrid Moeller; Trisha A. Jenkins; Siew Yeen Chai; Andrew M. Allen; Mitsuru Ohishi; Frederick A.O. Mendelsohn

Background The renin–angiotensin system (RAS) functions as both a circulating endocrine system and a tissue paracrine/autocrine system. As a circulating peptide, angiotensin II (Ang II) plays a prominent role in blood-pressure control and body fluid and electrolyte balance by acting on the AT1 receptor in the brain and peripheral tissues. As a paracrine/autocrine peptide, locally formed Ang II also plays additional roles in tissues involving the regulation of regional haemodynamics, cell growth and remodelling, and neurotransmitter release. Evidence is emerging that Ang II is not the only active peptide of the RAS, and other Ang II fragments may also have important biological activities. Objectives To provide a morphological basis for understanding novel actions of angiotensin-converting enzyme (ACE), Ang II and related peptides in tissues, this article will review the localization of ACE and AT1, AT2 and AT4 receptors in the central nervous system, blood vessels and kidney. Results and conclusion Autoradiographic mapping of the major components of the RAS has proved a valuable strategy to reveal, or suggest, cellular sites of novel actions for Ang II and related peptides in tissues. First, colocalization of ACE and AT1 receptors in the substantia nigra, the caudate nucleus and putamen of human and rat brain, which contain the dopamine-synthesizing neurons, suggests that the central RAS may be important in modulating central dopamine release. Secondly, the distribution of AT4 receptors with a striking association with cholinergic neurons, motor and sensory nuclei in the brain reveals that Ang IV may modulate central motor and sensory activities and memory. Thirdly, the occurrence of high levels of ACE and AT1 and/or AT2 receptors in the adventitia of blood vessels suggests important paracrine roles of the vascular RAS. Finally, the identification of abundant AT1 receptor and elucidation of its roles in the renomedullary interstitial cells of the kidney may provide a new impetus to study further the role of Ang II in the regulation of renal medullary function and blood pressure. Overall, circulating and locally produced Ang II and related peptides may exert a remarkable range of actions in the brain, kidney and cardiovascular system through multiple angiotensin receptors.


Neuroscience | 2004

Effect of I.C.V. injection of AT4 receptor ligands, NLE1-angiotensin IV and LVV-hemorphin 7, on spatial learning in rats

Joohyung Lee; Anthony L. Albiston; Andrew M. Allen; Frederick A.O. Mendelsohn; S.E Ping; Graham L. Barrett; Michael Murphy; Margaret J. Morris; Sharon G. McDowall; Syn Y Chai

Central administration of angiotensin IV (Ang IV) or its analogues enhance performance of rats in passive avoidance and spatial memory paradigms. The purpose of this study was to examine the effect of a single bolus injection of two distinct AT4 ligands, Nle1-Ang IV or LVV-haemorphin-7, on spatial learning in the Barnes circular maze. Mean number of days for rats treated with either Nle1-Ang IV or LVV-haemorphin-7 to achieve learner criterion is significantly reduced compared with controls (P < 0.001 and P < 0.05 respectively). This is due to enhanced ability of the peptide-treated rats to adopt a spatial strategy for finding the escape hatch. In all three measures of learning performance, (1) the number of errors made, (2) the distance travelled and (3) the latency in finding the escape hatch, rats treated with either 100 pmol or 1 nmol of Nle1-Ang IV or 100 pmol LVV-haemorphin-7 performed significantly better than the control groups. As early as the first day of testing, the rats treated with the lower dose of Nle1-Ang IV or LVV-haemorphin-7 made fewer errors (P < 0.01 and P < 0.05 respectively) and travelled shorter distances (P < 0.05 for both groups) than the control animals. The enhanced spatial learning induced by Nle1-Ang IV (100 pmol) was attenuated by the co-administration of the AT4 receptor antagonist, divalinal-Ang IV (10 nmol). Thus, administration of AT4 ligands results in an immediate potentiation of learning, which may be associated with facilitation of synaptic transmission and/or enhancement of acetylcholine release.

Collaboration


Dive into the Andrew M. Allen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael J. McKinley

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

George Paxinos

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robin M. McAllen

Florey Institute of Neuroscience and Mental Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge