The pathophysiology of advanced heart failure,☆☆,

https://doi.org/10.1016/S0002-8703(98)70252-2Get rights and content

Abstract

Am Heart J 1998;135:S216-S230.

Section snippets

Remodeling

The failing heart is accompanied by a change in ventricular shape and dimension, a process known as remodeling. Remodeling may be regional or global, depending on the type of insult to ventricular function (i.e., regional infarction vs cardiomyopathy) and is brought about by an increase in myocardial mass, an increase in ventricular volume, a change in ventricular shape, and interstitial growth. The dysfunctioning heart compromises stroke volume, and one adaptive compensatory mechanism to

Mechanical Considerations

Patients with advanced heart failure have for the most part enlarged ventricles, which place them at several mechanical disadvantages. The increased radius increases wall stress by Laplace's law, which is incompletely compensated for by the increase in wall thickness that occurs. In addition, the heart operates in a flat portion of the Starling mechanism. Although cardiac output and ventricular performance may be normal at rest, during exercise a marked elevation in pulmonary pressure may occur

Coronary Artery Disease

Heart failure in the setting of coronary artery disease is a heterogeneous condition with several factors contributing to left ventricular dysfunction and heart failure. These include AMI with loss of functional myocytes, development of myocardial fibrosis, and ventricular remodeling. In addition, the presence of coronary disease gives rise to the conditions of stunning and hibernation.

Myocardial stunning is defined as delayed recovery of myocardial function despite restoration of coronary

Arrhythmogenesis

Sudden death accounts for 28% to 68% of all deaths in patients with advanced heart failure treated with angiotensin-converting enzyme inhibitors.47, 48 In patients with relatively well-compensated heart failure, ventricular fibrillation may occur as a primary electrical event such as reentry in a chronic infarct scar; it can also occur in patients with very severe left ventricular dysfunction and hemodynamics in whom preventing or promptly terminating the arrhythmia is unlikely to alter the

Subendocardial Ischemia

Unverferth et al.52 raised the hypothesis that patients with primary cardiomyopathy are stranded in a self-perpetuating course of advancing heart failure. Among other factors, the progressive deterioration is caused by insufficient coronary blood flow to the subendocardium with a resultant loss of the area's ability to contribute to the work of the heart, initiating a vicious cycle that worsens the heart failure and further decreases coronary blood flow.56 Myocardial oxygen demand in patients

Molecular Changes

In advanced heart failure several molecular abnormalities have been identified, affecting in particular contractile protein function and excitation-contraction coupling. These have been demonstrated to be caused by a variety of mechanical, neurohumoral, and autocrine forces. Changes in contractile protein function appear to comprise an initial increase in protein production in response to ventricular overload, accompanied by a reversion to more fetal forms and an eventual reduction in protein

Myocyte Loss

In advanced cardiac failure ongoing myocyte loss is a feature of the myopathic process6, 68 and may occur by either necrosis or apoptosis. β-adrenergic stimulation including cardiac norepinephrine release and exposure to angiotensin II14can produce myocyte necrosis in model systems. In addition, aldosterone69, 70 has been shown to produce myocyte necrosis in rats after prolonged therapy (3 weeks). In this model spironolactone was protective, as were potassium supplements, indicating that the

Endothelial Dysfunction, Skeletal Muscle Dysfunction, and Cardiac Cathexia

In severe heart failure factors other than activity of the sympathetic nervous system and the RAS contribute to peripheral vasoconstriction. α-blockade and angiotensin-converting enzyme inhibition do not restore normal blood flow during exercise.78 Processes that depend on nitrous oxide (NO) such as acetylcholine-mediated vasodilatation and flow-dependent vasodilatation have been shown to be impaired.79, 80 In addition, response to glyceryl trinitrate is impaired. Endothelial dysfunction of

Neurohumoral Changes

Several complementary mechanistic pathways contribute to the physiological adjustments necessary for optimum cardiac function in health and disease. After an insult to cardiac function occurs, however, these mechanisms may cause a paradoxical worsening of cardiac function.

The sympathetic nervous system and the renin-angiotensin system constitute the primary neurohumoral systems for compensating a failing heart, causing largely an increase in afterload and preload. Many other pathways, presently

Role of Cellular Immune Activation

Cytokines are small polypeptides produced by a variety of immune cells and essentially represent an intercellular signaling system. The system is complicated; individual cytokines may be produced by more than one type of cell and may stimulate or inhibit more than one type of cell (Table I).91, 92Often one episode of cytokine release will have multiple effects, but most of the effect is in the locality of release, so that these molecules are largely a paracrine signaling system.

Role of Neurohumoral Pathways

A myriad of peptide signaling systems exist within the body, but in recent years attention has focused on two systems that have been demonstrated to be intimate with the appearance of congestive heart failure: the natriuretic peptides and the endothelins (Figure 3).

. Elevation of plasma ANP and BNP accompany severe heart failure and are related to the degree of LV dysfunction. Similar elevation of iNOS is seen in humans with severe heart failure. These initially homeostatic mechanisms do seem,

Conclusions

Severe heart failure causes the activation of compensatory immunologic and neurohumoral mechanisms. Although perhaps designed to be physiologically beneficial, these mechanisms may be wholly insufficient or, indeed, may paradoxically worsen the cardiac decompensation, creating opportunities for new therapeutic interventions. Simultaneously, the new identification of these plasma constituents may allow more accurate risk stratification of individuals at risk of ventricular dysfunction and

References (164)

  • T Kono et al.

    Left ventricular shape is the primary determinant of functional mitral regurgitation in heart failure

    J Am Coll Cardiol

    (1992)
  • LW Stevenson et al.

    Importance of hemodynamic response to therapy in predicting survival with ejection fraction less than or equal to 20% secondary to ischemic or nonischemic dilated cardiomyopathy

    Am J Cardiol

    (1990)
  • MO Jeroudi et al.

    Prolonged wall motion abnormalities after chest pain at rest in patients with unstable angina: a possible manifestation of myocardial stunning

    Am Heart J

    (1994)
  • Y Takeishi et al.

    Functional recovery of hibernating myocardium after coronary bypass surgery: does it coincide with improvement in perfusion?

    Am Heart J

    (1991)
  • M Cohen et al.

    Reversal of chronic ischemic myocardial dysfunction after transluminal coronary angioplasty

    J Am Coll Cardiol

    (1988)
  • E Downar et al.

    Endocardial mapping of ventricular tachycardia in the intact human heart: II. Evidence for multiuse reentry in a function sheet of surviving myocardium

    J Am Coll Cardiol

    (1992)
  • D Unverferth et al.

    The role of subendocardial ischaemia in perpetuating myocardial failure in patients with non-ischaemic congestive cardiomyopathy

    Am Heart J

    (1983)
  • M Packer et al.

    Provocation of hyper and hypokalemia sudden death during treatment with and withdrawal of converting-enzyme inhibition in severe chronic congestive heart failure

    Am J Cardiol

    (1986)
  • K Gallagher et al.

    Subepicardial vasodilator reserve in the presence of critical coronary stenosis in dogs

    Am J Cardiol

    (1980)
  • MJ Arends et al.

    Apoptosis: mechanisms and roles in pathology

    Internat Rev Exp Pathol

    (1991)
  • H Drexler et al.

    Endothelial function in congestive heart failure

    Am Heart J

    (1993)
  • T Inoue et al.

    Vasodilatory capacity of coronary resistance vessels in dilated cardiomyopathy

    Am Heart J

    (1994)
  • M Otaki

    Surgical treatment of patients with cardiac cachexia. An analysis of factors affecting operative mortality

    Chest

    (1994)
  • DL Mann et al.

    Basic mechanisms in congestive heart failure. Recognizing the role of proinflammatory cytokines

    Chest

    (1994)
  • J Lahdevirta et al.

    Elevated levels of circulating cachectin/tumor necrosis factor in patients with acquired immunodeficiency syndrome

    Am J Med

    (1988)
  • F Balkwill et al.

    Evidence for tumour necrosis factor/cachectin production in cancer

    Lancet

    (1987)
  • W Colucci et al.

    Pathophysiology of heart failure

  • MA Pfeffer et al.

    Ventricular remodeling after myocardial infarction. Experimental observations and clinical implications

    Circulation

    (1990)
  • F Messerli

    Pathophysiology of left ventricular hypertrophy

  • JB Caulfield et al.

    The collagen network of the heart

    Lab Invest

    (1979)
  • KT Weber et al.

    Structural remodelling of the heart by fibrous tissue: role of circulating hormones and locally produced peptides

    Eur Heart J

    (1995)
  • CA Beltrami et al.

    Structural basis of end-stage failure in ischemic cardiomyopathy in humans

    Circulation

    (1994)
  • CG Brilla et al.

    Remodelling of rat right and left ventricles in experimental hypertension

    Circ Res

    (1990)
  • D Mann et al.

    Load reduction of the properties of adult feline cardiac myocytes: growth induction by cellular deformation

    Circ Res

    (1989)
  • LB Tan et al.

    Cardiac myocyte necrosis induced by angiotensin II

    Circ Res

    (1991)
  • CG Brilla et al.

    Regulation of the structural remodelling of the myocardium: from hypertrophy to heart failure

    Eur Heart J

    (1994)
  • H Ito et al.

    Endothelin-1 induces hypertrophy with enhanced expression of muscle-specific genes in cultured neonatal rat cardiomyocytes

    Circ Res

    (1991)
  • JF Aceto et al.

    [Sar1]angiotensin II receptor-mediated stimulation of protein synthesis in chick heart cells

    Am J Physiol

    (1990)
  • KM Baker et al.

    Renin-angiotensin system involvement in pressure-overload cardiac hypertrophy in rats

    Am J Physiol

    (1990)
  • KM Baker et al.

    Angiotensin II stimulation of protein synthesis and cell growth in chick heart cells

    Am J Physiol

    (1990)
  • TG Parker et al.

    Growth factors, proto-oncogenes, and plasticity of the cardiac phenotype

    Ann Rev Physiol

    (1991)
  • TG Parker et al.

    Peptide growth factors can provoke “fetal” contractile protein gene expression in rat cardiac myocytes

    J Clin Invest

    (1990)
  • CG Brilla et al.

    Impaired diastolic function and coronary reserve in genetic hypertension. Role of interstitial fibrosis and medial thickening of intramyocardial coronary arteries

    Circ Res

    (1991)
  • KM Borow et al.

    Sensitivity of end-systolic pressure-dimension and pressure-volume relations to the inotropic state in humans

    Circulation

    (1982)
  • JK Perloff et al.

    The mitral apparatus. Functional anatomy of mitral regurgitation

    Circulation

    (1972)
  • B Patel et al.

    Postischemic myocardial “stunning”: a clinically relevant phenomenon

    Ann Intern Med

    (1988)
  • E Braunwald et al.

    The stunned myocardium: prolonged, postischemic ventricular dysfunction

    Circulation

    (1982)
  • GR Heyndrickx et al.

    Regional myocardial functional and electrophysiological alterations after brief coronary artery occlusion in conscious dogs

    J Clin Invest

    (1975)
  • S Zimmer et al.

    Metabolic correlates of reversibly injured myocardium

  • R Bolli

    Mechanism of myocardial “stunning.”

    Circulation

    (1990)
  • Cited by (0)

    From the aDepartment of Cardiological Sciences, St. George's Hospital Medical School, London; the bDivision of Cardiology, Northwestern University Medical School, Chicago; and the cDepartment of Cardiology, Cleveland Clinic Foundation.

    ☆☆

    Reprint requests: M. Kamran Baig, MD, Department of Cardiological Sciences, St. George's Hospital Medical School, Cranmer Terrace, London, UK SW17 ORE.

    4/0/90084

    View full text