309
Views
26
CrossRef citations to date
0
Altmetric
Review

Hypertension and other morbidities with Cushing’s syndrome associated with corticosteroids: a review

, &
Pages 7-16 | Published online: 03 Mar 2011

Abstract

Corticosteroids constitute an ideal treatment for various inflammatory and autoimmune disorders due to their anti-inflammatory and immunomodulatory actions. However, corticosteroids have a considerable number of side effects, including hypertension, diabetes, lipid disorders, sleep apnea, osteoporosis, myopathy, and disorders of coagulation and fibrinolysis, which are components of Cushing’s syndrome (CS). Corticosteroid-induced side effects are dependent on the formulation, route, dose, and time of exposure. However, the underlying pathogenetic mechanisms have not been clearly defined. A large body of evidence supports the role of an imbalance between vasoconstriction and vasodilation with possible links to nitric oxide, prostanoids, angiotensin II, arginine vasopressin, endothelins, catecholamines, neuropeptide Y, and atrial natriuretic peptide. Increased oxidative stress, renin–angiotensin system activation, increased pressor response, metabolic syndrome, and sleep apnea appear to be pathogenetically involved as well. The ideal treatment is the withdrawal of corticosteroids, which is most often impossible due to the exacerbation of the underlying disease. Alternatively, a careful plan, including the proper selection of the formulation, time, and route, should be made, and each side effect should be treated properly. The focus of the research should be to develop synthetic corticosteroids with anti-inflammatory effects but fewer metabolic effects, which so far has been unsuccessful.

Introduction

It is estimated that up to 0.5% of the US population are on chronic corticosteroid therapy for various inflammatory and autoimmune disorders.Citation1,Citation2 Annually, ∼10 million new prescriptions for oral corticosteroids are issued in the US. A minority of patients use corticosteroids without being aware of this, due to their use in the black market. Corticosteroids have been used, along with hydroquinone and mercury, as ingredients in a variety of skin lightening (bleaching) cosmetics and toiletries that are widely used in African countries.Citation3 Over-the-counter combination preparations of steroids with other drugs such as antifungals may lead to unsupervised and inappropriate use of topical corticosteroids.Citation2,Citation4,Citation5 Cases of factitious Cushing’s syndrome (CS) from surreptitious use of corticosteroids have been described.Citation6 Corticosteroids as an ingredient of a black market drug used by addicts to help them through the narcotic withdrawal stage were responsible for an outbreak of CS in Tehran in 2008.Citation7

Corticosteroids have been the medication of choice in various disorders, based on their undoubted benefits from their anti-inflammatory and immunomodulatory actions. However, they are also complicated by a considerable number of side effects, including CS. The clinical presentation of CS often strikes with the use of high doses of corticosteroids.Citation1,Citation2 Symptoms include central obesity, plethora, easy bruising, thin skin, striae, myopathy, depression or psychosis, poor wound healing, increased incidence of infection, glaucoma and other ocular disease, and hypertension (HT). Hirsutism and other virilizing features are uncommon clinical findings due to nonsignificant increases in androgens. Avascular necrosis and spinal epidural lipomatosis constitute a complication of corticosteroid-induced CS.Citation1,Citation2

All synthetic derivatives that are used clinically, including prednisone, prednisolone, methylprednisolone, dexamethasone (DX), betamethasone, and triamcinolone, have the potential for adverse effects and CS.Citation2 Which of these agents is most likely to cause CS has not been fully clarified due to the complex interaction of many factors involved in this process.Citation2 It appears that it is dependent on the formulation used, pharmacokinetics, affinity for the glucocorticoid receptor, biologic potency, duration of action, and different levels of sensitivity in individual patients.Citation2 Oral corticosteroid therapy has been well correlated with CS, and most physicians are aware of the dangers, having always to balance the cost–effect ratio.Citation2 According to accumulated data, topical, aerosol, inhaled, and injectable corticosteroid therapy may also have adverse effects, including CS.Citation2,Citation8Citation12 There have been several cases of children who received intralesional injections into keloid scars or other wounds (such as burns) and developed CS that persisted for a long time (up to 9 months).Citation13 Cases of CS have also been reported in patients under treatment with paraspinal depot injections.Citation14 Additional unusual cases include CS induced by serial occipital nerve blocks containing triamcinolone.Citation15 Corticosteroid-related side effects including CS are common in patients with cystic fibrosis or HIV infection, which need combined treatments including budesonide and itraconazole or fluticasone propionate and ritonavir.Citation11,Citation12,Citation16,Citation17

Another critical determinant of the corticosteroid-induced side effects including CS is the dosage needed to control the disease. High doses of corticosteroids even for a short time or long-term use of agents with lower potency and short half-lives (hydrocortisone and cortisone) have been associated with CS. However, the prediction of dosages at which CS will develop is a complicated phenomenon that depends on a variety of factors.Citation1,Citation2 Thus, the cost–effect ratio has to be estimated in each case, and a careful plan should be made before starting treatment with corticosteroids. This review will discuss the critical determinants and underlying pathogenetic mechanisms of CS associated with corticosteroids.

Morbidities associated with corticosteroid-related CS

Corticosteroid-induced CS and HT

HT is a prominent feature in patients with corticosteroid-induced CS, occurring in up to 20% of cases, and is dose dependent.Citation1,Citation2,Citation18,Citation19 A variety of mechanisms has been proposed to explain its pathogenesis.Citation18,Citation19

A large body of evidence supports the theory that corticosteroids induce an imbalance between vasoconstriction and vasodilation, favoring vasoconstriction, resulting in HT.Citation18,Citation19 According to in vitro, in vivo, and also human data, increased vasoconstriction is in large part mediated by increased endothelin-1 synthesis and secretion.Citation20Citation22 Increased cytosolic calcium levels downregulate the expression of the Na–Ca exchanger and increased erythropoeitin levels have also been pathogenetically involved in corticosteroid-induced vasoconstriction and HT.Citation23,Citation24 Accumulating data suggest that increased sympathetic activity, reflected in the increased synthesis of catecholamines and a1β-adrenergic receptor expression, is an important underlying pathogenetic mechanism as well.Citation18,Citation19 The increased synthesis of cat-echolamines is due to increased expression and activity of various enzymes involved in the catecholamine biosynthesis, including tyrosine hydroxylase and phenylethanolamine N-methyltransferase. Elevated epinephrine and norepinephrine levels in plasma and adrenal medulla and increased expression and activity of tyrosine hydroxylase have been found in hypertensive rats treated with subcutaneous injections of dexamethasone (DX; 1 mg/kg/day for 2 days).Citation25 Elevated levels of plasma dopamine and epinephrine have been found in humans treated with a single dose of 2 mg DX.Citation26 In both studies, the observed effects were blocked by the administration of alpha-methyl-p-tyrosine, an inhibitor of tyrosine hydroxylase.Citation25,Citation26 Increased activity of phenyle-thanolamine N-methyltransferase has been found in both intact and adrenalectomized rats treated with subcutaneous injections of DX (1 mg/kg/day for 12–14 days).Citation27,Citation28 In addition, in vitro, in vivo, and human studies suggest that corticosteroids alter the availability of α1-adrenergic receptors in vascular smooth muscles, leading to increased vascular reactivity, pressor responsiveness, and HT.Citation29Citation31

Increased corticosteroid-induced vasoconstriction and thus HT are also mediated through enhanced synthesis and action of vasoactive substances and their receptors, including neuropeptide Y (NPY), arginine vasopressin (AVP), and atrial natriuretic peptide (ANP).Citation19,Citation20 Increased corticosteroid-induced NPY gene expression and tissue content in neuroendocrine tissue and cell lines have been associated with vasoconstriction.Citation32 Coadministration of AVP and DX (1.8 mg/kg/week for 2 weeks) to normotensive rats resulted in dose-dependent increases in mean arterial pressure, which can be reversed by the administration of d(CH2)5Tyr(Me) AVP, an AVP-V1 receptor antagonist, suggesting the role of the V1aAVP receptor in DX-induced HT.Citation31,Citation33,Citation34 Increased expression of atrial, ventricular, pulmonary ANP genes and plasma ANP levels have been found in both intact and adrenalectomized rats treated with DX (1 and 4 mg/kg/day for 2 days).Citation31,Citation35 Higher ANP levels have been observed in atrial slices and extracts obtained from rats treated with DX (intraperitoneally, 0.1 mg/kg/day for 4 days).Citation36,Citation37

Another interesting issue is the role of renin–angiotensin system activation in the development of corticosteroid-induced HT.Citation18,Citation19 Corticosteroids act directly at the liver site, enhancing the synthesis of the plasma renin substrate (angiotensinogen).Citation31,Citation38 Experimental studies using saralasin, an angiotensin antagonist, and SQ14225, an angiotensin-converting enzyme, partly prevented HT in rats treated with DX (0.17–0.27 mg/kg/day), suggesting the partial contribution of angiotensin II in terms of increased synthesis or sensitivity in the development of DX-induced HT.Citation38

A large body of evidence supports the theory that apart from the increased corticosteroid-induced synthesis and secretion of vasoactive substances, an increased sensitivity and reactivity of various tissues to their action, reflected in increased vascular pressor responsiveness, has also been observed.Citation31,Citation39Citation41 A reduced threshold and increased maximal response to norepinephrine has been observed in rat mesenteric vasculatures isolated from hypertensive rats treated with DX orally (7–9.5 mg/kg/day for 28 days).Citation42,Citation43

AVP, but not norepinephrine or angiotensin II infusion, elicited increased pressor response in DX-induced hypertensive rats (257 mg/kg/day for 2 weeks).Citation33 Infusion of angiotensin II in humans treated with DX (orally, 3 mg/day) resulted in increased forearm vascular resistance.Citation39,Citation44 These effects seem to be mediated through changes in the activity of sodium/potassium pumpCitation45 and function of glucocorticoidCitation46 and mineralocorticoidCitation47 receptors as well. Administration of angiotensin II and AVP in DX-treated experimental animals was accompanied by a reduction in the threshold of the inositol triphosphate production and HT, an effect that was blocked by the administration of a specific glucocorticoid receptor antagonist RU38486 but not spironolactone or RU28318, type I mineralocorticoid receptor antagonists.Citation46,Citation48 DX-induced stimulation of vascular angiotensin II type 1 receptor has also been observed and linked to HT.Citation47 Furthermore, corticosteroids seem to induce HT by binding to mineralocorticoid receptors.Citation49 It has been supported that although corticosteroids activate both mineralocorticoid and glucocorticoid receptors, they exhibit a higher affinity for glucocorticoid receptors than with mineralocorticoid receptors.Citation50

On the other hand, corticosteroids negatively affect various vasodilatory systems.Citation18,Citation19 Corticosteroid-induced HT has been associated with nitric oxide (NO) deficiency through a range of negative influences on the NO biosynthetic pathways, involving i) alteration in the activity and expression of NO synthase, ii) decreased availability of tetrahydrobiopterin (BH4), and iii) decreased NO precursor l-arginine.Citation51Citation53 NO deficiency might also be the result of NO interaction with excess superoxide to form a powerful oxidant, peroxynitrite, which leads to NO inactivation and deficit.Citation54,Citation55

According to experimental data, corticosteroids seem to negatively affect the production of other vasodilatory substances as well, such as prostacyclin, prostaglandin E2 (PGE2), and kallikrein.Citation56Citation58

The end result of the afore mentioned alterations is hemodynamic changes in various vascular beds. A large dose of oral DX (0.5 mg/kg/day) in dogs was accompanied by a reduction in cardiac output and an increase in calculated total peripheral resistance.Citation31 In humans, oral DX (1 mg three times daily for 7 days) increased mean and total peripheral vascular resistance without affecting the cardiac output.Citation39 Limited studies support an effect of DX on the regional hemodynamics as well. Intravenous 24 h infusion of DX (125 mg/kg/h) increased the mean arterial pressure, and decreased renal and mesenteric blood flow and conductance.Citation59 However, it remains unclear whether this is a coexisting feature or a pathogenic mechanism of corticosteroid-induced HT.

Coexisting metabolic abnormalities appear to mediate and accentuate the corticosteroid-induced HT. Obesity is associated with a reduction in urine sodium excretion, increased plasma and extracellular fluid volume, and HT.Citation18,Citation19 Insulin resistance leads to sodium and water retention, increased sympathoadrenal system activity, local renin–angiotensin system activation, vascular hypertrophy, increased vascular resistance, and HT.Citation18,Citation19 Sleep apnea has also been associated with HT through increased sympathetic tone during hypoxemic episodes, insulin resistance, and diabetic autonomic neuropathy.Citation18,Citation19

It seems that corticosteroids regulate blood pressure by exerting their effects at the central nervous system as well. Subcutaneous tritium-labeled DX administration in rats resulted in localization of radioisotope in the thalamus (lateral nucleus), hypothalamus (arcuate, ventromedial, periventricular, and paraventricular nuclei), and cell bodies of locus ceruleus, area postrema, and nucleus tractus solitarii, indicating that systemically administered DX passes to the brain and cerebrospinal fluid.Citation60Citation62 An interesting theory has been developed suggesting that corticosteroids induce central nervous system activation and HT, but this has to be further evaluated.Citation62 This effect might be mediated through direct interaction of corticosteroids with γ-aminobutyric acid type A and B receptors and nontranscriptional activation of phosphatidylinositol 3-kinase/protein kinase Akt pathway, possibly mediated by the glucocorticoid receptor.

Corticosteroid-induced CS and osteoporosis

Osteoporosis constitutes an important component of corticosteroid-induced CS in children and adults.Citation1,Citation2 Bone loss appears to be fastest in the first 6 months of therapy and persists at a slower rate thereafter, resulting in osteopenia and osteoporosis. Trabecular bone and the cortical rim of vertebral bodies appear to be more susceptible to the effects of corticosteroids, improving rapidly after their withdrawal. In addition, fractures may occur in 30%–50% of corticosteroid-treated patients independently of bone mineral density differences.Citation63,Citation64 Long-term use of corticosteroids in asthmatic patients has been associated with increased rib and vertebral fractures.Citation65 A retrospective cohort study of 244,235 adults on oral corticosteroids documented a dose-dependent increase in nonvertebral, hip, forearm, and vertebral fractures, occurring even with low doses of corticosteroids (2.5 mg/day).Citation66 A meta-analysis of 42,500 men and women from seven prospective cohorts showed that current or prior corticosteroid use has been associated with increased fracture risk.Citation67 There is a large body of evidence indicating that inhaled corticosteroids negatively affect bone metabolism, as well as when they are taken orally.Citation68,Citation69 However, the percentage of bone loss and the real fracture risk attributed to corticosteroids is difficult to be estimated in those patients, due to the multifactorial origin of altered bone metabolism.

Corticosteroids negatively affect calcium metabolism and bone remodeling, leading to decreased bone formation and increased bone resorption. Corticosteroids decrease intestinal calcium absorption and renal tubular reabsorption of calcium, resulting in hypercalciuria, secondary hyperparathyroidism, and increased bone resorption.Citation63,Citation64 Corticosteroids have a negative impact on osteoblastogenesis as well by reducing synthesis of type 1 collagen and insulin-like growth factor, altering the binding and thus the anabolic effects of transforming growth factor-β, inhibiting Wnt signaling, and inducing apoptosis of osteoblasts and osteocytes.Citation63,Citation64 Decreased activity of type 1-11β-hydroxysteroid dehydrogenase and increased activity of type 2–11β-hydroxysteroid dehydrogenase by circulating inflammatory cytokines, the antagonistic effect of corticosteroids on parathormone and testosterone, seem to represent underlying pathogenetic mechanisms. On the other hand, corticosteroids stimulate osteoclast proliferation by suppressing synthesis of osteoprotegerin, stimulating production of the receptor activator of nuclear factor κB and decreasing estrogen and androgen production, leading to increased osteoclastic bone resorption.Citation63,Citation64,Citation70Citation72

Corticosteroid-induced CS and disorders of the coagulation/fibrinolysis system

Thromboembolic disease is an important and serious complication in patients with CS, as it is associated with increased morbidity and mortality.Citation1,Citation2,Citation73 Small clinical studies in patients with CS have shown an activation of the coagulation system reflected in higher levels of plasma von Willebrand factor VIII, factor IX, and factor XII; decreased fibrinolytic activity reflected in lower levels of PAI-1, tPA, or euglobulin clot lysis time; and increased levels of factors XII, XI, IX, and VIII plasminogen and a 2-antiplasmin.Citation74,Citation75 These effects have been closely related to corticosteroids, as they were fully reversed after surgical treatment. In a similar way, exogenous corticosteroids cause disorders in the coagulation and fibrinolytic systems, reflected in increased synthesis/secretion of PAI-1, and increased levels of thrombin–antithrombin complex increased plasma factor VII, factor VIII, factor XI, and fibrinogen levels.Citation76Citation78

Corticosteroid-induced CS and muscloskeletal disorders

Myopathy seems to be a common feature in CS, clinically expressed as the inability to rise from a crouching position, due to the adverse effects of corticosteroids on the proximal muscles of the lower limb and the shoulder girdle.Citation1,Citation2,Citation79 Corticosteroids affect type 2B or ‘phasic’ muscle fibers (fast twitch) causing atrophy (but not necrosis), resulting in reduced muscle protein synthesis. This effect is mediated through increased oxidative phosphorylation, inhibition of protein synthesis, and impairment of muscle membrane excitability.Citation79

The acute form of myopathy involves proximal and distal muscle weakness, is positively correlated with elevated serum creatinine phosphokinase levels, is an indicator of focal and diffuse muscle necrosis, is mediated by hypokalemia, or constitutes the end result of a direct effect of corticosteroids on skeletal muscle. This type of myopathy may take between 6 weeks and several months to resolve, even after the discontinuation of corticosteroids. The chronic form is more insidious in onset, primarily involves proximal muscle groups, and is characterized by typically normal or only slightly elevated creatinine phosphokinase levels with no evidence of focal or diffuse muscle necrosis. Although there is no direct correlation with dosage, patients who receive even small amounts of prednisone (ie, 10 mg/day) can develop myopathy.Citation80,Citation81

Corticosteroid-induced CS and metabolic syndrome

Impaired glucose tolerance, diabetes, dyslipidemia, and fatty liver disease are common findings in patients treated with corticosteroids.Citation82Citation84 Not only experimental but also human data support the theory of corticosteroids having an effect on various sites involved in protein, lipid, and glucose metabolism, including skeletal muscle, liver, and adipose tissue. Corticosteroids induce insulin resistance in skeletal muscle by directly interfering with the insulin signaling cascade. Decreased glucogen synthesis rates and glycogen synthase concentration and activity have been found in skeletal muscle biopsies of healthy subjects on 4 mg DX for 4 daysCitation85 and patients on long-term treatment with high doses of corticosteroids following renal transplantation.Citation86 Skeletal muscle insulin resistance is further aggravated by corticosteroid-induced protein catabolism with concomitant atrophy-related decrease in total muscle area and elevated circulating aminoacids,Citation87 which negatively affect insulin signaling,Citation88 glucose uptake, and glycogen synthesis in muscle.Citation89 In addition, corticosteroids induce whole body lipolysis,Citation89 resulting in increased plasma levels of free fatty acids and triglyceridesCitation90,Citation91 with deleterious effects in skeletal muscle insulin sensitivity and glucose uptake. Corticosteroids induce hepatic insulin resistance as well, directly by interference with insulin signaling and indirectly by elevating free fatty acid and triglyceride supply to the liver.Citation92 Thus, increased endogenous glucose production has been observed, especially in the postprandial state, as was demonstrated in healthy subjects following short-term exposure to corticosteroids.Citation84,Citation93,Citation94 Corticosteroids increase body fat content and alter body fat distribution by regulating hormone sensitive lipase and lipoprotein lipase activity.Citation95 In addition, corticosteroids modulate adipose tissue biology by altering the secretion of adipokines either directly or through insulin resistance.Citation96 In addition to inducing insulin resistance, corticosteroids might exert an inhibitory effect on β cells, which is dependent on duration of exposure, dosage, and susceptibility of the population exposed. A prolonged exposure (2–5 days) of healthy subjects to high doses of corticosteroids resulted in fasting hyperinsulinemia and increased insulin secretion, possibly to compensate for the corticosteroid-induced insulin resistance.Citation97Citation99 However, in a susceptible population, such as people with a reduced insulin sensitivity before treatment, people who are healthy first-degree relatives of patients with diabetes, and obese women, this compensation failed, resulting in β cell failure and diabetes.Citation88,Citation98Citation100 Thus, corticosteroids induce insulin resistance, impaired glucose metabolism, obesity, and central obesity, all of which are components of metabolic syndrome, which has been associated with increased morbidity and mortality from cardiovascular disease.

Corticosteroid-induced CS and cardiovascular disease

Patients with corticosteroid-induced CS exhibit increased cardiovascular morbidity and mortality.Citation1,Citation2,Citation101Citation103 Increased mortality from cardiovascular disease has been reported in patients with asthma, chronic obstructive pulmonary disease, inflammatory arthritis, and giant cell arteritis.Citation104Citation106 A population-based study showed that patients who were exposed to systematic corticosteroids in a dose greater than the equivalent of 7.5 mg of prednisolone daily had substantially higher rates of cardiovascular disease during 1–5 years of follow-up, namely myocardial infarction, heart failure, and cerebrovascular disease.Citation102,Citation103 It has to be mentioned that this association was not evident in patients treated with low doses or ‘nonsystemic’ (eg, topical and rectal) corticosteroids.

Corticosteroids appear to have a direct causal effect relationship with cardiovascular disease, which is dependent on the dose, duration, cumulative dose of exposure, and route of administration. Whether, and to what extent, the increased risk is mediated through the disease background for cardiovascular disease and the induction of several risk factors for cardiovascular disease and represents a direct corticosteroid effect that is much higher than their anti-inflammatory and antiproliferative actions cannot be answered by the existing data.Citation107,Citation108

Corticosteroid-induced CS and fetal programming

Much interest has been focused over the last 5 years on the role of corticosteroids in fetal programming.Citation1,Citation2,Citation109 The key mediators appear to be the hypothalamic–pituitary–adrenal axis, the glucocorticoid receptor, and the expression of type 2–11β-hydroxysteroid dehydrogenase genes in a range of tissues. The administration of corticosteroids to mothers to promote maturation of organs in fetuses, to prevent a number of life-threatening complications of pre-term birth, and to reduce the effects of congenital adrenal hyperplasia has undoubted short-term benefits and also potential long-term adverse effects.Citation109,Citation110 A large body of experimental and human evidence supports the theory that corticosteroids appear to be involved in a dose-dependent manner in the fetal programming of adult diseases.Citation111 Corticosteroid-related fetal programming of adult HT is exerted through the effects on maturation of tissues involved in the control of blood pressure, such as glomerular number and kidney size, in the expression of catecholamine receptors and second messenger systems in renal and vascular tissue, and by affecting growth factors and carbohydrate and fat homeostasis. In addition, corticosteroids potentiate vasoconstrictor effects on the vasculature and regulate the synthesis of catecholamines, NO, and angiotensinogen.Citation112,Citation113 HT is coupled to tissue-specific increases in glucocorticoid receptor expression and downregulation of type 2–11β-hydroxysteroid dehydrogenase activity in the placenta, kidney, and adrenal, increasing sensitivity and overexposing organs to corticosteroids. Apart from inducing HT, corti-costeroids are involved in the fetal programming of type 2 diabetes, cardiovascular disease, other manifestations of the metabolic syndrome, several central nervous system functions, and psychiatric syndromes. However, the exact underlying pathogenetic mechanisms are still unclear and need to be further elucidated.

Exogenous vs endogenous corticosteroid-induced CS and HT

Although the clinical presentation of CS is quite similar from endogenous cortisol overproduction and exogenous corticosteroids, it also has some differences, mainly in the more striking clinical manifestations, due to the use of high doses of corticosteroids.Citation1,Citation2 CS as a result of the long-term usage of corticosteroids has less hirsutism and other virilizing features due to nonsignificant increases in androgens. In addition, HT is less profound in exogenous corticosteroid-induced CS compared with endogenous CS, depending on the form used. In contrast, avascular necrosis and spinal epidural lipomatosis occur primarily in the setting of corticosteroid-induced CS.Citation1,Citation2 In addition, a kinetic difference has to be mentioned. In contrast to cortisol, most synthetic corticosteroids bind to albumin and not to cortisol-binding globulin or circulate as free steroids with a much higher affinity for the glucocorticoid receptor. With HT, both forms of CS exhibit some similarities and some differences.

In both forms of CS, the development of HT is rapid. Supraphysiological oral cortisol doses (80 and 200 mg/day) in humans can cause HT within 24 h, with peak blood pressure occurring at day 4 or 5 of treatment.Citation114,Citation115 In addition, subcutaneous DX (10 mg/day) in rats and oral DX (0.5 mg/kg/day) in dogs and humans (3 mg/day) increases blood pressure within 1–2 days.Citation31 According to in vitro and in vivo studies, the same pattern is observed in endogenous CS as well.Citation116,Citation117 HT, in both clinical entities, is independent of miner-alocorticoid activity and sodium loading or retention.Citation49,Citation118 However, in endogenous CS, sodium excess can magnify the hypertensive response.Citation119,Citation120 Increased oxidative stress, reflected in increased superoxide production and elevated levels of plasma F2-isoprostanes, constitutes a common underlying disorder in both forms of CS and is prevented and reversed by antioxidants (folic acid, N-acetylcysteine, tempol, and apocynin) but not with BH4 or allopurinol.Citation121Citation123 In both conditions, HT has been associated with decreased NO bioavailability, reflected in decreased plasma reactive nitrogen intermediates (nitrate/nitrite).Citation122,Citation123

Although HT, in both forms of CS, is associated with increased oxidative stress and NO deficiency and inactivation, blood pressure response to treatments known to modify the synthesis of NO and superoxide seems to be variable. l-Arginine treatment (500 mg/kg/day) increased plasma nitrate/nitrite concentrations but failed to prevent HT in corticosteroid-treated rats.Citation53,Citation121 In contrast, in endogenous CS, HT was prevented and partly reversed by l-arginine treatment.Citation124 Aspirin, an antioxidant and nonselective cyclooxygenase inhibitor, prevented and partly reversed HT related to endogenous but not exogenous CS.Citation125 Vasopressin antagonism with an AVP-V1 receptor antagonist significantly decreased mean arterial pressure in exogenous but not endogenous CS-related hypertensive rats.Citation33 Both forms of HT exhibit a different response to glucocorticoid receptor antagonism, suggesting a different degree of receptor activation in both situations. Thus, dehydroepiandroster-one, an endogenous steroid with antiglucocorticoid activity, prevented corticosteroid-induced but not endogenous cortisol-induced HT.Citation31,Citation126 In summary, it appears that HT due to endogenous or exogenous CS involves either different pathophysiological mechanisms or different degree of perturbations of the same mechanisms.

Treatment

Patients with autoimmune or inflammatory disorders should be on long-term use of corticosteroids and may develop HT and other morbidities associated with corticosteroid-induced CS. The ideal treatment is the withdrawal of corticosteroids before the onset of comorbidities. However, this is practically impossible, as it is associated with exacerbation of the underlying disease. When possible, corticosteroids should be gradually withdrawn. The use of corticosteroids in low dosages or on alternative days results in fewer metabolic effects and also helps in the prevention of corticosteroid-related pathology. Thus, each patient should be treated individually, following a careful, properly designed plan. In most cases, patients should be treated for corticosteroid-related morbidities. For HT, eplerenone, angiotensin receptor antagonists, and angiotensin-converting enzyme inhibitors are indicated. Weight loss is encouraged through changes in the lifestyle. Medications that improve insulin resistance, such as biguanides and peroxisome proliferator-activated receptor agonists, should be added. Treatment of sleep apnea with a continuous positive airway pressure device appears to be generally effective in controlling not only apnea but also HT, insulin resistance, and increased risk for cardiovascular disease. Bisphosphonates are the first-line choice for prevention or treatment of osteoporosis, with teriparatide as the second-line option; calcium and vitamin D supplements should be coprescribed in the majority of individuals. Muscle biopsy is recommended in patients who require long-term corti-costeroid treatment and develop weakness during therapy. Phenytoin has been suggested for treatment of corticosteroid myopathy, but this still needs further research. In the absence of prospective randomized clinical trials, there is currently general agreement that patients with CS should be treated as having a prothrombotic disorder. However, future large prospective trials are needed to evaluate the type, intensity, and duration of thromboprophylaxis in patients with either endogenous CS or iatrogenic CS.

Conclusion

Corticosteroid-induced CS constitutes a major health problem with difficult handling. A careful plan for treatment, namely formulation, route, dose, and time of exposure, should be properly evaluated individually before patients start corticosteroids use. Corticosteroid-related complications should be treated properly, if evident, and prevention strategy involves changes in lifestyle and treatment with appropriate agents. The focus of the research should be on developing synthetic steroids with anti-inflammatory but fewer metabolic effects, which so far has been unsuccessful.

Disclosure

The authors report no conflicts of interest in this work.

References

  • PivonelloRde MartinoMCde LeoMLombardiGColaoACushing’s syndromeEndocrinol Metab Clin North Am2008371135149ix18226734
  • ChrousosGPGlucocorticoid therapyFeligPFrohmanLEndocrinology and Metabolism4th edNew YorkMcGraw-Hill Incorporated2001609632
  • OlumideYMAkinkugbeAOAltraideDComplications of chronic use of skin lightening cosmeticsInt J Dermatol200847434435318377596
  • Castanedo-CazaresJPLopez-LucioRHMoncadaBCushing syndrome following the prescription of antifungal, antibiotic, corticosteroid creamInt J Dermatol200342431812694505
  • WeberSLCushing’s syndrome attributable to topical use of lotrisoneEndocr Pract19973314014415251475
  • VillanuevaRBBrettEGabriloveJLA cluster of cases of factitious Cushing’s syndromeEndocr Pract20006214314711421530
  • AziziFJahedAHedayatiMOutbreak of exogenous Cushing’s syndrome due to unlicensed medicationsClin Endocrinol (Oxf)200869692192518462262
  • ErmisBOrsRTastekinAOzkanBCushing’s syndrome secondary to topical corticosteroids abuseClin Endocrinol (Oxf)200358679579612780758
  • LipworthBJSystemic adverse effects of inhaled corticosteroid therapy: a systematic review and meta-analysisArch Intern Med1999159994195510326936
  • PerryRJFindlayCADonaldsonMDCushing’s syndrome, growth impairment, and occult adrenal suppression associated with intranasal steroidsArch Dis Child2002871454812089123
  • KumarSSinghRJReedAMLteifANCushing’s syndrome after intra-articular and intradermal administration of triamcinolone acetonide in three pediatric patientsPediatrics200411361820182415173517
  • LansangMCFarmerTKennedyLDiagnosing the unrecognized systemic absorption of intra-articular and epidural steroid injectionsEndocr Pract200915322522819364690
  • TeelucksinghSBalkaranBGaneshmoorthiAArthurPProlonged childhood Cushing’s syndrome secondary to intralesional triamcinolone acetonideAnn Trop Paediatr2002221899111926057
  • EdmondsLCVanceMLHughesJMMorbidity from paraspinal depo corticosteroid injections for analgesia: Cushing’s syndrome and adrenal suppressionAnesth Analg19917268208222035867
  • LavinPJWorkmanRCushing syndrome induced by serial occipital nerve blocks containing corticosteroidsHeadache200141990290411703480
  • BollandMJBaggWThomasMGLucasJATicehurstRBlackPNCushing’s syndrome due to interaction between inhaled corticosteroids and itraconazoleAnn Pharmacother2004381464914742792
  • GuptaSKDubeMPExogenous Cushing syndrome mimicking human immunodeficiency virus lipodystrophyClin Infect Dis2002356E69E7112203188
  • SarutaTSuzukiHHandaMIgarashiYKondoKSenbaSMultiple factors contribute to the pathogenesis of hypertension in Cushing’s syndromeJ Clin Endocrinol Metab19866222752793510223
  • MagiakouMASmyrnakiPChrousosGPHypertension in Cushing’s syndromeBest Pract Res Clin Endocrinol Metab200620346748216980206
  • RoubertPViossatILonchamptMOEndothelin receptor regulation by endothelin synthesis in vascular smooth muscle cells: effects of dexamethasone and phosphoramidonJ Vasc Res19933031391448518331
  • KirilovGTomovaADakovskaLKumanovPShinkovAAlexandrovASElevated plasma endothelin as an additional cardiovascular risk factor in patients with Cushing’s syndromeEur J Endocrinol2003149654955314640996
  • BorcsokISchairerHUSommerUGlucocorticoids regulate the expression of the human osteoblastic endothelin A receptor geneJ Exp Med19981889156315739802968
  • SmithLSmithJBRegulation of sodium-calcium exchanger by glucocorticoids and growth factors in vascular smooth muscleJ Biol Chem19942694427527275317961668
  • MacefieldVGWilliamsonPMWilsonLRKellyJJGandeviaSCWhitworthJAMuscle sympathetic vasoconstrictor activity in hydrocortisone-induced hypertension in humansBlood Press1998742152229858113
  • KumaiTAsohKTateishiTInvolvement of tyrosine hydroxylase up regulation in dexamethasone-induced hypertension of ratsLife Sci200067161993199911072875
  • WatanabeTNoshiroTAkamaHEffect of dexamethasone on plasma free dopamine: dopaminergic modulation in hypertensive patientsHypertens Res199518Suppl 1S197S1988529061
  • KennedyBElayanHZieglerMGGlucocorticoid hypertension and nonadrenal phenylethanolamine N-methyltransferaseHypertension19932144154198458643
  • KennedyBZieglerMGCardiac epinephrine synthesis. Regulation by a glucocorticoidCirculation19918428918951860230
  • SakaueMHoffmanBBGlucocorticoids induce transcription and expression of the alpha 1B adrenergic receptor gene in DTT1 MF-2 smooth muscle cellsJ Clin Invest19918823853891650792
  • HaighRMJonesCTEffect of glucocorticoids on alpha 1-adrenergic receptor binding in rat vascular smooth muscleJ Mol Endocrinol19905141482168709
  • NakamotoHSuzukiHKageyamaYCharacterization of alterations of hemodynamics and neuroendocrine hormones in dexamethasone induced hypertension in dogsClin Exp Hypertens A19911345876061934541
  • EdvinssonLEkbladEHakansonRWahlestedtCNeuropeptide Y potentiates the effect of various vasoconstrictor agents on rabbit blood vesselsBr J Pharmacol19848325195256593107
  • IijimaFMalikKUContribution of vasopressin in dexamethasone-induced hypertension in ratsHypertension1988112 Pt 2I42I463346065
  • MurasawaSMatsubaraHKizimaKMaruyamaKMoriYInadaMGlucocorticoids regulate V1a vasopressin receptor expression by increasing mRNA stability in vascular smooth muscle cellsHypertension19952646656697558228
  • GardnerDGHaneSTrachewskyDSchenkDBaxterJDAtrial natriuretic peptide mRNA is regulated by glucocorticoids in vivoBiochem Biophys Res Commun19861393104710542945558
  • GarciaRDebinskiWGutkowskaJGluco- and mineralocorticoids may regulate the natriuretic effect and the synthesis and release of atrial natriuretic factor by the rat atria in vivoBiochem Biophys Res Commun198513128068142932106
  • LachanceDGarciaRGutkowskaJCantinMThibaultGMechanisms of release of atrial natriuretic factor. I. Effect of several agonists and steroids on its release by atrial mincesBiochem Biophys Res Commun19861353109010982938580
  • KlettCGantenDHellmannWRegulation of hepatic angiotensinogen synthesis and secretion by steroid hormonesEndocrinology19921306366036681597163
  • PirpirisMSudhirKYeungSJenningsGWhitworthJAPressor responsiveness in corticosteroid-induced hypertension in humansHypertension1992196 Pt 15675741592452
  • SambhiMPWeilMHUdhojiVNPressor responses to norepinephrine in humans before and after corticosteroidsAm J Physiol196220396196313991288
  • LeferAMManwaringJLVerrierRLEffect of corticosteroids on the cardiovascular responses to angiotensin and norepinephrineJ Pharmacol Exp Ther1966154183914288615
  • RussoDFraserRKenyonCJDexamethasone therapy selectively increases the sensitivity to noradrenaline of the rat mesenteric circulationJ Hypertens Suppl198976S126S1272632693
  • RussoDFraserRKenyonCJIncreased sensitivity to noradrenaline in glucocorticoid-treated rats: the effects of indomethacin and desipramineJ Hypertens1990898278332172372
  • GriendlingKKMinieriCAOllerenshawJDAlexanderRWAngiotensin II stimulates NADH and NADPH oxidase activity in cultured vascular smooth muscle cellsCirc Res1994746114111488187280
  • SternNPalantCOzakiLTuckMLDexamethasone enhances active cation transport in cultured aortic smooth muscle cellsAm J Hypertens1994721461508179849
  • SatoASuzukiHIwaitaYNakazatoYKatoHSarutaTPotentiation of inositol trisphosphate production by dexamethasoneHypertension19921911091151730435
  • SatoASuzukiHNakazatoYShibataHInagamiTSarutaTIncreased expression of vascular angiotensin II type 1A receptor gene in glucocorticoid-induced hypertensionJ Hypertens19941255115167930550
  • GrunfeldJPEloyLMouraAMGanevalDRamos-FrendoBWorcelMEffects of antiglucocorticoids on glucocorticoid hypertension in the ratHypertension1985722922993980072
  • WilliamsonPMKellyJJWhitworthJADose-response relationships and mineralocorticoid activity in cortisol-induced hypertension in humansJ Hypertens Suppl1996145S37S419120683
  • RebuffatAGTamSNawrockiARThe 11-ketosteroid 11-ketodexamethasone is a glucocorticoid receptor agonistMol Cell Endocrinol20042141–2273715062542
  • WallerathTWitteKSchaferSCDown-regulation of the expression of endothelial NO synthase is likely to contribute to glucocorticoid-mediated hypertensionProc Natl Acad Sci U S A19999623133571336210557325
  • SimmonsWWUngureanu-LongroisDSmithGKSmithTWKellyRAGlucocorticoids regulate inducible nitric oxide synthase by inhibiting tetrahydrobiopterin synthesis and L-arginine transportJ Biol Chem19962713923928239378798625
  • LiMFraserTWangJWhitworthJADexamethasone-induced hypertension in the rat: effects of L-arginineClin Exp Pharmacol Physiol1997249–107307329315378
  • PacherPBeckmanJSLiaudetLNitric oxide and peroxynitrite in health and diseasePhysiol Rev200787131542417237348
  • IuchiTAkaikeMMitsuiTGlucocorticoid excess induces superoxide production in vascular endothelial cells and elicits vascular endothelial dysfunctionCirc Res2003921818712522124
  • ZhangYHuLMoriTABardenACroftKDWhitworthJAArachidonic acid metabolism in glucocorticoid-induced hypertensionClin Exp Pharmacol Physiol2008355–655756218067589
  • FalardeauPMartineauAProstaglandin I2 and glucocorticoid-induced rise in arterial pressure in the ratJ Hypertens1989786256322509553
  • NasjlettiAErmanACagenLMBaerPGPlasma concentrations, renal excretion, and tissue release of prostaglandins in the rat with dexamethasone-induced hypertensionEndocrinology19841143103310406421564
  • GardinerSMKempPAMarchJEBennettTEffects of dexamethasone and SB 209670 on the regional haemodynamic responses to lipopolysaccharide in conscious ratsBr J Pharmacol199611811411498733587
  • SiegalTSotiFBiegonAPopEBrewsterMEEffect of a chemical delivery system for dexamethasone (Dex-CDS) on peritumoral edema in an experimental brain tumor modelPharm Res19971456726759165542
  • BirminghamMKSarMStumpfWEDexamethasone target sites in the central nervous system and their potential relevance to mental illnessCell Mol Neurobiol19931343733868252608
  • WangLLOuCCChanJYReceptor-independent activation of GABAergic neurotransmission and receptor-dependent nontranscriptional activation of phosphatidylinositol 3-kinase/protein kinase Akt pathway in short-term cardiovascular actions of dexamethasone at the nucleus tractus solitarii of the ratMol Pharmacol200567248949815523051
  • GravesLLukertBPGlucocorticoid-induced osteoporosis: a clinician’s perspectiveClin Rev Bone Miner Metab2004227990
  • ShakerJLLukertBPOsteoporosis associated with excess glucocorticoidsEndocrinol Metab Clin North Am2005342341356viiiix15850846
  • AdinoffADHollisterJRSteroid-induced fractures and bone loss in patients with asthmaN Engl J Med198330952652686866051
  • Van StaaTPLeufkensHGAbenhaimLZhangBCooperCUse of oral corticosteroids and risk of fracturesJ Bone Miner Res2000156993100010841167
  • KanisJAJohanssonHOdenAA meta-analysis of prior corticosteroid use and fracture riskJ Bone Miner Res200419689389915125788
  • WongCAWalshLJSmithCJPInhaled corticosteroid use and bone-mineral density in patients with asthmaLancet200035592131399140310791523
  • Van StaaTPLeufkensHGCooperCUse of inhaled corticosteroids and risk of fracturesJ Bone Miner Res200116358158811277277
  • CooperMSRabbittEHGoddardPEBartlettWAHewisonMStewartPMOsteoblastic 11beta-hydroxysteroid dehydrogenase type 1 activity increases with age and glucocorticoid exposureJ Bone Miner Res200217697998612054173
  • WeinsteinRSJilkaRLParfittAMManolagasSCInhibition of osteoblastogenesis and promotion of apoptosis of osteoblasts and osteocytes by glucocorticoids. Potential mechanisms of their deleterious effects on boneJ Clin Invest199810222742829664068
  • OdellWDTestosterone treatment of men treated with glucocorticoids (editorial)Arch Intern Med199615611113311348639006
  • La BroccaATerzoloMPiaAPaccottiPde GiuliPAngeliARecurrent thromboembolism as a hallmark of Cushing’s syndromeJ Endocrinol Invest19972042112149211128
  • EremCNuhogluIYilmazMBlood coagulation and fibrinolysis in patients with Cushing’s syndrome: increased plasminogen activator inhibitor-1, decreased tissue factor pathway inhibitor, and unchanged thrombin-activatable fibrinolysis inhibitor levelsJ Endocrinol Invest200932216917419411818
  • PatrassiGMDal Bo ZanonRBoscaroMMartinelliSGirolamiAFurther studies on the hypercoagulable state of patients with Cushing’s syndromeThromb Haemost19855425185204082087
  • OikarinenAHoythyaMJarvinenMDexamethasone-induced plasminogen activator inhibitor: characterization, purification, and preparation of monoclonal antibodiesArch Dermatol Res199028231531582142402
  • Barouski-MillerPAGelehrterTDParadoxical effects of glucocorticoids on regulation of plasminogen activator activity of rat hepatoma cellsPNAS1982797231923226179095
  • BrotmanDJGirodJPPoschAEffects of short-term glucocorticoids on hemostatic factors in healthy volunteersThromb Res2006118224725216005496
  • HasselgrenPOGlucocorticooids and muscle catabolismCurr Opin Clin Nutr Metab Care19992320120510456248
  • WilliamsTJO’HehirRECzarnyDHorneMBowesGAcute myopathy in severe acute asthma treated with intravenously administered corticosteroidsAm Rev Respir Dis198813724604633341634
  • Van MarleWWoodsKLAcute hydrocortisone myopathyBr Med J198028162352712727427239
  • LarssonHAhrenBInsulin resistant subjects lack islet adaptation to short-term dexamethasone-induced reduction in insulin sensitivityDiabetologia199942893694310491753
  • HenriksenJEAlfordFWardGMBeck-NielsenHRisk and mechanism of dexamethasone-induced deterioration of glucose tolerance in non-diabetic first-degree relatives of NIDDM patientsDiabetologia19974012143914489447952
  • RizzaRAMandarinoLJGerichJECortisol-induced insulin resistance in man: impaired suppression of glucose production and stimulation of glucose utilization due to a postreceptor detect of insulin actionJ Clin Endocrinol Metab19825411311387033265
  • HenriksenJEAlfordFVaagAHandbergABeck-NielsenHIntracellular skeletal muscle glucose metabolism is differentially altered by dexamethasone treatment of normoglycemic relatives of type 2 diabetic patientsMetabolism19994891128113510484052
  • EkstrandASchalin-JanttiCLofmanMThe effect of (steroid) immunosuppression on skeletal muscle glycogen metabolism in patients after kidney transplantationTransplantation19966168898938623155
  • SchakmanOGilsonHThissenJPMechanisms of glucocorticoid-induced myopathyJ Endocrinol2008197111018372227
  • KrebsMKrssakMBernroiderEMechanism of amino acid-induced skeletal muscle insulin resistance in humansDiabetes200251359960511872656
  • DinneenSAlzaidAMilesJRizzaREffects of the normal nocturnal rise in cortisol on carbohydrate and fat metabolism in IDDMAm J Physiol19952684 Pt 1E595E6037733257
  • TaskinenMRNikkilaEAPelkonenRSaneTPlasma lipoproteins, lipolytic enzymes, and very low density lipoprotein triglyceride turnover in Cushing’s syndromeJ Clin Endocrinol Metab19835736196266348067
  • WajchenbergBLSubcutaneous and visceral adipose tissue: their relation to the metabolic syndromeEndocr Rev200021669773811133069
  • ParekhSAnaniaFAAbnormal lipid and glucose metabolism in obesity: implications for nonalcoholic fatty liver diseaseGastroenterology200713262191220717498512
  • NielsenMFCaumoAChandramouliVImpaired basal glucose effectiveness but unaltered fasting glucose release and gluconeogenesis during short-term hypercortisolemia in healthy subjectsAm J Physiol Endocrinol Metab20042861E102E11012965873
  • RooneyDPNeelyRDCullenCThe effect of cortisol on glucose/glucose-6-phosphate cycle activity and insulin actionJ Clin Endocrinol Metab1993775118011838077310
  • MortonNMSecklJR11beta-hydroxysteroid dehydrogenase type 1 and obesityFront Horm Res20083614616418230901
  • FasshauerMPaschkeRRegulation of adipocytokines and insulin resistanceDiabetologia200346121594160314605806
  • BeardJCHalterJBBestJDPfeiferMAPorteDJrDexamethasone-induced insulin resistance enhances B cell responsiveness to glucose level in normal menAm J Physiol19842475 Pt 1E592E5966388353
  • GrillVPigonJHartlingSGBinderCEfendicSEffects of dexamethasone on glucose-induced insulin and proinsulin release in low and high insulin respondersMetabolism19903932512582407926
  • MatsumotoKYamasakiHAkazawaSHigh-dose but not low-dose dexamethasone impairs glucose tolerance by inducing compensatory failure of pancreatic beta-cells in normal menJ Clin Endocrinol Metab1996817262126268675587
  • BesseCNicodNTappyLChanges in insulin secretion and glucose metabolism induced by dexamethasone in lean and obese femalesObes Res200513230631115800288
  • ManciniTKolaBManteroFBoscaroMArnaldiGHigh cardiovascular risk in patients with Cushing’s syndrome according to 1999 WHO/ISH guidelinesClin Endocrinol (Oxf)200461676877715579193
  • WeiLMacDonaldTMWalkerBRTaking glucocorticoids by prescription is associated with subsequent cardiovascular diseaseAnn Intern Med20041411076477015545676
  • SouvereinPCBerardAvan StaaTPUse of oral glucocorticoids and risk of cardiovascular and cerebrovascular disease in a population based case-control studyHeart200490885986515253953
  • HoleDJWattGCDavey-SmithGHartCLGillisCRHawthorneVMImpaired lung function and mortality risk in men and women: findings from the Renfrew and Paisley prospective population studyBMJ199631370597117158819439
  • DeMariaANRelative risk of cardiovascular events in patients with rheumatoid arthritisAm J Cardiol.2002896A33D38D
  • UddhammarAErikssonALNystromLStenlingRRantapaa-DahlqvistSIncreased mortality due to cardiovascular disease in patients with giant cell arteritis in northern SwedenJ Rheumatol200229473774211950015
  • NashelDJIs atherosclerosis a complication of long-term corticosteroid treatment?Am J Med19868059259293518440
  • MaxwellSRMootsRJKendallMJCorticosteroids: do they damage the cardiovascular system?Postgrad Med J1994708308638707870631
  • BertramCEHansonMAPrenatal programming of postnatal endocrine responses by glucocorticoidsReproduction2002124445946712361463
  • KayHHBirdIMCoeCLDudleyDJAntenatal steroid treatment and adverse fetal effects: what is the evidence?J Soc Gynecol Investig200075269278
  • NewnhamJPIs prenatal glucocorticoid administration another origin of adult disease?Clin Exp Pharmacol Physiol2001281195796111703405
  • Langley-EvansSCShermanRCWelhamSJNwagwuMOGardnerDSJacksonAAIntrauterine programming of hypertension: the role of the renin-angiotensin systemBiochem Soc Trans1999272889310093713
  • BenediktssonRLindsayRNobleJSecklJREdwardsCRGlucocorticoid exposure in utero: new model for adult hypertensionLancet199334188413393418094115
  • ConnellJMWhitworthJADaviesDLLeverAFRichardsAMFraserREffects of ACTH and cortisol administration on blood pressure, electrolyte metabolism, atrial natriuretic peptide and renal function in normal manJ Hypertens1987544254332822795
  • KellyJJTamSHWilliamsonPMLawsonJWhitworthJAThe nitric oxide system and cortisol-induced hypertension in humansClin Exp Pharmacol Physiol199825119459469807669
  • TurnerSWWenCLiMFraserTBWhitworthJAAdrenocorticotrophin dose-response relationships in the rat: haemodynamic, metabolic and hormonal effectsJ Hypertens19981655936009797170
  • WhitworthJAAdrenocorticotrophin and steroid-induced hypertension in humansKidney Int Suppl199237S34S371321309
  • MillsEHCoghlanJPDentonDASpenceCDWhitworthJAScogginsBAThe effect of sodium depletion and potassium loading on cortisol induced hypertension in sheepActa Endocrinol (Copenh)198611322983043776438
  • WhitworthJASainesDScogginsBAPotentiation of ACTH hypertension in man with salt loadingClin Exp Pharmacol Physiol19851232392432992854
  • HumphreyTJFanJSCoghlanJPInter-relationships between sodium and potassium intake and the blood pressure effects of ACTH in sheepJ Hypertens19831119266099379
  • HuLZhangYLimPSApocynin but not L-arginine prevents and reverses dexamethasone-induced hypertension in the ratAm J Hypertens200619441341816580579
  • MondoCKZhangYde Macedo PossamaiVN-acetylcysteine antagonizes the development but does not reverse ACTH-induced hypertension in the ratClin Exp Hypertens2006282738416546835
  • ZhangYChanMMAndrewsMCApocynin but not allopurinol prevents and reverses adrenocorticotropic hormone-induced hypertension in the ratAm J Hypertens200518791091616053986
  • WenCLiMFraserTWangJTurnerSWWhitworthJAL-arginine partially reverses established adrenocorticotrophin-induced hypertension and nitric oxide deficiency in the ratBlood Press20009529830411193135
  • ZhangYMiaoYWhitworthJAAspirin prevents and partially reverses adrenocorticotropic hormone-induced hypertension in the ratAm J Hypertens200720111222122817954371
  • LiMWenCMartinAWhitworthJADehydroepiandrosterone does not prevent adrenocorticotrophin-induced hypertension in conscious ratsClin Exp Pharmacol Physiol19962354354378713685