Term
|
Definition
|
|
Term
|
Definition
|
|
Term
|
Definition
|
|
Term
|
Definition
|
|
Term
|
Definition
|
|
Term
CLINICAL MANIFESTATIONS. The clinical manifestations of ARDS are progressive as follows: |
|
Definition
Dyspnea and hypoxemia with poor response to oxygen supplementation
Hyperventilation and respiratory alkalosis
Decreased tissue perfusion, metabolic acidosis, and organ dysfunction
Increased work of breathing, decreased tidal volume, and hypoventilation
Hypercapnia, respiratory acidosis, and worsening hypoxemia Decreased cardiac output, hypotension, and death |
|
|
Term
|
Definition
(Systolic BP 160 mmHg or diastolic BP 100 mmHg) Two-drug combination for most (usually thiazide-type diuretic and ACE inhibitor or ARB or beta-blocker or CCB) |
|
|
Term
|
Definition
Lymphocytes with receptors against self antigens are eliminated |
|
|
Term
|
Definition
these cells are suppress by regulatory T-cells and other mechanism |
|
|
Term
|
Definition
Antibody producing cell- T-regulatory cells that suppress the immune response to self-antigen |
|
|
Term
|
Definition
small antigen that binds to large molecule and induces an immune response |
|
|
Term
|
Definition
portion of the antigen that is recognized and bound by an antibody or specific lymphocyte. Antigenic determinant |
|
|
Term
|
Definition
molecule that activates many the cell receptors regardless of their antigen specificity by binding in an usual location |
|
|
Term
|
Definition
Presents endogenous antigen |
|
|
Term
|
Definition
presents exogenous antigen |
|
|
Term
|
Definition
a substance included in a vaccine to stimulate immune response |
|
|
Term
|
Definition
Lymphocytes with receptors against self antigens are eliminated |
|
|
Term
|
Definition
these cells are suppress by regulatory T-cells and other mechanism |
|
|
Term
|
Definition
Antibody producing cell- T-regulatory cells that suppress the immune response to self-antigen |
|
|
Term
|
Definition
small antigen that binds to large molecule and induces an immune response |
|
|
Term
|
Definition
portion of the antigen that is recognized and bound by an antibody or specific lymphocyte. Antigenic determinant |
|
|
Term
|
Definition
molecule that activates many the cell receptors regardless of their antigen specificity by binding in an usual location |
|
|
Term
|
Definition
Presents endogenous antigen |
|
|
Term
|
Definition
presents exogenous antigen |
|
|
Term
|
Definition
a substance included in a vaccine to stimulate immune response |
|
|
Term
CD1 antigen-presenting molecules present exogenous___________ antigen |
|
Definition
|
|
Term
Helper T cells are MHC Class ___________ restricted, but cytoxi c cells are ___________MHC Class Restricted |
|
Definition
|
|
Term
The secondary immune response also is called the ___________ response |
|
Definition
|
|
Term
Cytokines secreted by the cell promote ___________ immunity; cytokines secreted by the cells promote ___________ immunity; cytokines secreted by th17 promote ___________; cytokines secreted by trey cells ___________ the immune response |
|
Definition
Cellular; humoral; inflammatory; suppress |
|
|
Term
class switch indicates that a b lymphoctye changes from producing ___________ to producing another class of IG |
|
Definition
|
|
Term
Cytotoxic T cell kill their target by secreting ___________that make a pore in the membrane and injecting ___________ through the pore, causing apoptosis |
|
Definition
|
|
Term
th1 cells activate macrophages by secreting___________ |
|
Definition
|
|
Term
|
Definition
Define against parasites; degrade vasoactive substance released by mast cells. |
|
|
Term
|
Definition
release chemicals that initiate the inflammatory response |
|
|
Term
|
Definition
Eliminate virus-infected cells |
|
|
Term
|
Definition
Phagocytize microorganism and cellular debris;secrete chemicals that promote tissue healing; activate adaptive immunity |
|
|
Term
|
Definition
the first line of deference against microorganism |
|
|
Term
Surfactants and other chemical defenses produced by lung epithelium are called ___________ |
|
Definition
|
|
Term
|
Definition
microorganism that normally colonized the body surfaces |
|
|
Term
pattern recognization receptors |
|
Definition
one innate immune cell can recognize many different type of pathogenic bacteria via this process |
|
|
Term
MAC is formed by activated___________cascade causes ___________ |
|
Definition
|
|
Term
List the cells that are phagocyte innate |
|
Definition
|
|
Term
List the cells that are non phagocytic innate |
|
Definition
Mast, Natural killer cells |
|
|
Term
List the cells that are adaptive |
|
Definition
|
|
Term
|
Definition
Brief vasoconstriction, Vasodilation, Increased vascular permeability, Leakage of palm into tissue, Local edema |
|
|
Term
Sequence the events that occur when a neutrophil phagocytes a microorganism in the tissue |
|
Definition
Increased adhesion molecules, margination, diapedesis, cemotaxix, recognition and attachment, engulfment and formation of phagosome, formation of phagolysosome, destruction of microorganism |
|
|
Term
|
Definition
is a pattern molecule associated with a pathogenic organisms |
|
|
Term
|
Definition
is a molecular pattern that is associated with injured or stressed host cells. |
|
|
Term
|
Definition
is nonspecific, meaning that one cell or other element will defend against many different type of antigens |
|
|
Term
|
Definition
is specific meaning that one cell will defend against only one particular antigen. |
|
|
Term
Sequence the events that occur when a neutrophil phagocytes a microorganism in the tissue |
|
Definition
Increased adhesion molecules, margination, diapedesis, cemotaxix, recognition and attachment, engulfment and formation of phagosome, formation of phagolysosome, destruction of microorganism |
|
|
Term
|
Definition
is a pattern molecule associated with a pathogenic organisms |
|
|
Term
|
Definition
is a molecular pattern that is associated with injured or stressed host cells. |
|
|
Term
|
Definition
is nonspecific, meaning that one cell or other element will defend against many different type of antigens |
|
|
Term
|
Definition
is specific meaning that one cell will defend against only one particular antigen. |
|
|
Term
|
Definition
are molecules that mark antigens for destruction by innate immune cells |
|
|
Term
|
Definition
are signaling molecules that influence behavior of immune and other type of cells. |
|
|
Term
|
Definition
The phagocyte leaves the bloodstream and enters the tissue through interactions between leukocyte and endothelial adhesion molecules and the process of diapedesis. |
|
|
Term
In a healing wound, fibroblast, migrate to the area, which phagocytic cell secretes chemicals to attract them |
|
Definition
|
|
Term
What is the function of the fibroblast |
|
Definition
To secrete collagen to help scar tissue |
|
|
Term
Does wound contraction occur before or after fibroblast migration and proliferation |
|
Definition
|
|
Term
Another word for natural immunity and native immunity |
|
Definition
|
|
Term
|
Definition
|
|
Term
|
Definition
|
|
Term
|
Definition
containing many red blood cells |
|
|
Term
|
Definition
Another term for purulent exudate |
|
|
Term
|
Definition
watery with few proteins or cells |
|
|
Term
|
Definition
meaning that original tissue structure and function have been restored if little tissue has been lost or injured tissue is capable of regeneration. |
|
|
Term
|
Definition
involve extensive damage or tissues incapable of regeneration heal by the process of repair that results in the formation of a scar. |
|
|
Term
The two stages of Resolution healing |
|
Definition
the reconstructive phase, in which the wound begins to heal, and the maturation phase, in which the healed wound is remodeled. |
|
|
Term
Order or development of mature CD4 cell |
|
Definition
Lymphoid stem cell migrate to thymus, cell divides and differentiates developing t-cell receptors and surface markers, immunocompetent naive tell migrate to secondary lymphoid organs, APC presents processed antigen with MHC class II molecule to Tcellm cell differentiate and matures to CD4 cells. |
|
|
Term
The Fab fragment binds to what |
|
Definition
antigen, it consists of two chains. |
|
|
Term
another name for an antigenic determinant |
|
Definition
|
|
Term
|
Definition
active against parasites are important mediator of allergies |
|
|
Term
|
Definition
Produced during the primary response to antigen, are the largest Igs |
|
|
Term
|
Definition
Are the most abundant class of Igs are transported across the placenta |
|
|
Term
|
Definition
has subclasses; one subclass is most abundant in body secretions |
|
|
Term
|
Definition
function unknown in blood are surface receptors on developing b lymphocytes |
|
|
Term
|
Definition
Plasma cell is the cell that makes antibodies; Antibodes are a protective part of immune system and normally defends us against bacteria and viruses. |
|
|
Term
|
Definition
an altered immunologic response to an antigen that results in disease or damage to the host |
|
|
Term
|
Definition
is a disturbance in the immunologic tolerance of self-antigens. |
|
|
Term
|
Definition
occurs when the immune system of one individual produces an immunologic reaction against tissues of another individual. Alloimmunity can be observed during immunologic reactions against transfusions, transplanted tissue, or the fetus during pregnancy. |
|
|
Term
Type I: IgE-Mediated Hypersensitivity Reactions |
|
Definition
Most common allergies (e.g., pollen allergies) are type I reactions. In addition, most type I reactions occur against environmental antigens and are therefore allergic. |
|
|
Term
II Tissue-specific reaction |
|
Definition
IgG IgM Macrophages in tissues Frequently Autoimmune thrombocytopenic purpura, Graves disease, autoimmune hemolytic anemia |
|
|
Term
III Immune complex– mediated reaction |
|
Definition
IgG IgM Neutrophils Yes Systemic lupus erythematosus |
|
|
Term
|
Definition
None Lymphocytes, macrophages No Contact sensitivity to poison ivy and metals (jewelry) |
|
|
Term
|
Definition
Steady state of tolerance to self-antigens or lack of immune reactions against environmental antigen. |
|
|
Term
|
Definition
immune system of one individual produces an immunologic reaction against tissues of another individual |
|
|
Term
Type II hypersensitivity is induced by 5 different mechanisms name them |
|
Definition
1. complement mediated lysis 2. opsonization and phagocytosis 3. neutrophil mediated tissue damage 4. antibody cell mediated toxicity 5. modulation of cellular function. |
|
|
Term
Complement mediated lysis occurs when |
|
Definition
IgG or IgM antibodies damage the cell membrane with resulting lysis of the cell membrane of the erythrocyte. This is seen in autoimmune hemolytic anemia or as a result of an ABO compatibility |
|
|
Term
Opsonization and phagocytosis |
|
Definition
occurs when opsonin bind to a membrane and phagocyte are attracted resulting removal by phagocytosis. |
|
|
Term
Neutrophil mediated tissue damage |
|
Definition
occurs through the attraction of neutrophils which release granules |
|
|
Term
Antibody cell-mediated toxicity |
|
Definition
ocuurs when ctyoxic cells which are not antigen specific release toxic substance to destry target cells |
|
|
Term
Modulation of cellular function |
|
Definition
The cell is not destroy but caused to malfunction. A perfect example of this is in Graves disease where autoantibodies bind to and activates receptors for TSH, thus resulting in production too much thyroxine and the failure of the negative feed back system |
|
|
Term
|
Definition
produced by macrophages and lymphocytes in response to the presence of an invading microorganism or activation of the inflammatory process. |
|
|
Term
|
Definition
are cytokines that primarily protect the host against viral infections and play a role in the modulation of the inflammatory response. I |
|
|
Term
|
Definition
mediated by the B lymphocytes (B cells) and is the principal defense against extracellular microbes and their toxins. The B cells differentiate into antibody- secreting plasma cells. The circulating antibodies then interact with and destroy the microbes that are present in the blood or mucosal surfaces |
|
|
Term
Cell-mediated, or cellular, immunity |
|
Definition
is mediated by the cytotoxic T lymphocytes (T cells) and functions in the elimination of intracellular pathogens (e.g., viruses). T cells develop receptors that recognize the viral peptides displayed on the surface of infected cells and then signal destruction of the infected cells. |
|
|
Term
The cytokines involved in innate immunity include |
|
Definition
TNF-α and lymphotoxin; interferons (IFN-γ, IFN-α, IFN-β); the interleukins IL-1, IL-6, and IL-12; and chemokines (see Table 13.2). These substances modulate innate immunity by stimulating the development of cells involved in both innate and adaptive immunity, producing chemotaxis within leukocytes, stimulating acute-phase protein production, and inhibiting viral replication |
|
|
Term
Two acute-phase proteins that are involved in the defense against infections |
|
Definition
mannose-binding ligand (MBL) and C-reactive protein (CRP). MBL and CRP are produced in the liver in response to activation of proinflammatory cytokines. MBL binds specifically to mannose residues, and CRP binds to both phospholipids and sugars that are found on the surface of microbes. These substances act as “costimulatory” opsonins and enhance the binding of phagocytic cells to suboptimally opsonized invading microorganisms.25 They |
|
|
Term
Regulatory T cells (TR) are a subset of T lymphocytes that function to control immune system responses |
|
Definition
These cells represent a subset of CD4+ cells that act as “negative regulators” of the immune process34. They suppress immune responses by inhibiting the proliferation of other potentially harmful self-reactive lymphocytes. |
|
|
Term
The primary function of cytotoxic T (CD8+) cells |
|
Definition
to monitor the activity of all cells in the body and destroy any that threaten the integrity of the body. CD8+ T cells recognize antigens that are presented on the cell surface by MHC class I–derived molecules that sample peptides from protein degradation productions from inside cells infected by viruses or transformed by cancer33 |
|
|
Term
|
Definition
the loss of cellular differentiation, irregularities of the size and shape of the nucleus, and the loss of normal tissue structure. |
|
|
Term
|
Definition
Cancers arising from connective tissu |
|
|
Term
|
Definition
to preinvasive epithelial malignant tumors of glandular or squamous cell origin. Cancers develop incrementally, as they accumulate specific genetic lesions. Careful surveillance for cancer often detects abnormal growths in epithelial tissues that have atypical cells and an increased proliferation rate compared with normal surrounding tissue |
|
|
Term
|
Definition
are of endocrine, neurologic, and/or immunologic etiology. These syndromes are a result of hormonal production of ectopic peptide by the tumor or from autoantibodies released in response to the tumor.9 |
|
|
Term
18F-Fluorodeoxyglucose (FDG) |
|
Definition
Because it is missing a key hydroxyl group it cannot be broken down by glycolysis and, thus, FDG accumulates in cells. Because it is tagged with 18F, it can be imaged by positron emission tomography (a PET scan) |
|
|
Term
|
Definition
for example, anchorage-independent growth, lack of contact inhibition, and immortality. |
|
|
Term
clonal proliferation or clonal expansion |
|
Definition
That mutant cell may then have a selective advantage over its neighbors; its progeny can accumulate faster than its nonmutant neighbors |
|
|
Term
|
Definition
promote cancer proliferation |
|
|
Term
|
Definition
are mutant genes that in their normal nonmutant state direct synthesis of proteins that positively regulate (accelerate) proliferatio |
|
|
Term
|
Definition
encode proteins that in their normal state negatively regulate (halt, or “put the brakes on”) proliferation. Hence, they also have been referred to as anti-oncogenes. |
|
|
Term
|
Definition
An example of a proto-oncogene would be a growth factor (e.g., epidermal growth factor) or a growth factor receptor (e.g., epidermal growth factor receptor). |
|
|
Term
|
Definition
|
|
Term
|
Definition
Cancers that arise because of these mutations often depend on these mutant genes and proteins for their continued growth and survival. |
|
|
Term
TYPES OF GENETIC LESIONS IN CANCER |
|
Definition
1. Point mutations 2. Subtle alterations (insertions, deletions) 3. Chromosome changes (aneuploidy and loss of heterozygosity) 4. Amplifications 5. Gene silencing (DNA methylation, histone modification, microRNAs) 6. Exogenous sequences (tumor viruses). Mary are found in as |
|
|
Term
|
Definition
the myc gene is aberrantly placed under the control of a B-cell immunoglobulin gene (Ig) present on chromosome 14. The Ig gene is very active in maturing B lymphocytes. The t(8;14) translocation alters the control of myc; its normal low level is switched to high levels, as directed by an Ig gene promoter. MYC protein, when inappropriately high, drives proliferation and blocks differentiation. |
|
|
Term
chronic myeloid leukemia (CML), |
|
Definition
This translocation, t(9;22),This translocation fuses two chromosomes in the middle of two different genes, bcr on chromosome 9 and abl on chromosome 22. The result is production of a BCR-ABL fusion protein containing the first half of BCR and the second half of ABL. BCR-ABL is a misregulated protein tyrosine kinase that promotes growth of myeloid cells Imatinib, a drug that specifically targets this tyrosine kinase, represents the first successful chemotherapy targeted against the product of a specific oncogenic mutation. Imatinib and related tyrosine kinase inhibitors (TKIs) are highly effective in the treatment of CML and, because of their specificity |
|
|
Term
|
Definition
Amplifications are the result of duplication of a small piece of a chromosome over and over again, so that instead of the normal two copies of a gene, tens or even hundreds of copies are presen |
|
|
Term
The N-myc oncogene is amplified in 25% of childhood neuroblastoma cases and confers a poor prognosis.17 |
|
Definition
Result in Gene Amplification |
|
|
Term
Gene amplification example |
|
Definition
The epidermal growth factor receptor erbB2 is amplified in 20% of breast cancers |
|
|
Term
|
Definition
are genes whose major function is to negatively regulate cell growth and prevent mutations. Tumor suppressors may normally slow the cell cycle, inhibit proliferation resulting from growth signals, or stop cell division when cells are damaged |
|
|
Term
|
Definition
a tumor suppressor gene, normally strongly inhibits the cell division cycle (see Chapter 1). When it is inactivated, the cell division cycle can proceed unchecked. Rb is mutated in childhood retinoblastoma, and in many lung, breast, and bone cancers as well. |
|
|
Term
|
Definition
involve changes in the patterns of gene expression without a change in the DNA. Epigenetic mechanisms may “silence” genes, such as tumor suppressor genes, so that even though the gene is present, it is not expressed and a cancer-suppressing protein is not made. Genes silenced by hypermethylation can be inherited, |
|
|
Term
Self-sufficiency in growth signa |
|
Definition
cancer cells stimulate their own growth; |
|
|
Term
Insensitivity to anti-growth signals |
|
Definition
They proliferate even though there are anti growth signals present. epidermal growth factor (EGF) receptor HER2/neu is up-regulated, and likely sends growth signals into the cell even when growth factors are at very low levels. Inhibitors of HER2 and other EGF receptors that block this pathway are effective in treating selected breast and lung cancers.34 |
|
|
Term
|
Definition
ability to secrete growth factors that stimulate their own growth. |
|
|
Term
Up to one third of all cancers have an activating mutation in the gene for an intracellular signaling protein called |
|
Definition
|
|
Term
|
Definition
More advanced cancers can, however, secrete multiple factors that stimulate new blood vessel growth (called neovascularization or angiogenesis). The angiogenic factors, such as vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), and basic fibroblast growth factor (bFGF), |
|
|
Term
|
Definition
monoclonal antibody that inhibits VEGF; erlotinib, sorafenib, and sunitinib, inhibitors of the VEGF and PDGF receptor tyrosine kinases; and thalidomide |
|
|
Term
tumor-associated macrophage, or TAM. |
|
Definition
Uses cytokines and cheekiness to attract monocytes or macrophages to the tumor, which The classical pro-inflammatory macrophage (M1) is the primary macrophage in the acute inflammatory response and is responsible for removal and destruction of infectious agents. During healing, however, a different phenotype (M2) produces anti-inflammatory mediators to suppress ongoing inflammation and induce cellular proliferation, angiogenesis, and wound healing.64 TAMs appear to phenotypically mimic the M2 phenotype. The transition to the M2 phenotype is under control of Th2 cytokines (for example, tumor necrosis factor-alpha [TNF-α], colony-stimulating factor-2 [CSF2; also known as granulocyte/macrophage colony stimulating factor or GM-CSF], interleukin |
|
|
Term
How does TAM uses our pathophysiology to keep the tumor alive |
|
Definition
TAMs have diminished cytotoxic response, and develop the capacity to block cytotoxic T-cell and NK cell functions and produce cytokines that are advantageous for tumor growth and spread. TAMs secrete cellular growth factors (for example, TGF-β and fibroblast growth factor [FGF-2]) that favor tumor cell proliferation, angiogenesis, |
|
|
Term
matrix metalloproteinases (MMPs) and plasminogen activators |
|
Definition
Active proteases digest the extracellular matrix and basement membranes, creating pathways through which cells can move can and travel to other areas. |
|
|
Term
(epithelial-mesenchymal transition [EMT]) |
|
Definition
increased migratory capacity, elevated resistance to apoptosis, and the potential to redifferentiate into other cell types. These properties permit the tumor cells to dissociate from the primary tumor and be transported to a distal site. |
|
|
Term
|
Definition
T0 = Breast free of tumor T1 = Lesion <2 cm in size T2 = Lesion 2-5 cm T3 = Skin and/or chest wall involved by invasion |
|
|
Term
|
Definition
N0 = No axillary nodes involved N1 = Mobile nodes involved N2 = Fixed nodes involved |
|
|
Term
|
Definition
M0 = No metastases M1 = Demonstrable metastases M2 = Suspected metastases |
|
|
Term
|
Definition
are symptom complexes that are triggered by a cancer but are not caused by direct local effects: They are most commonly caused by biologic substances released from the tumor (e.g., hormones) or by an immune response triggered by the tumor of the tumor mass |
|
|
Term
|
Definition
Parathyroid hormone–related protein (PTHRP), TGF-α, TNF, IL-1 |
|
|
Term
|
Definition
|
|
Term
|
Definition
RANK ORDER* CAUSE OF DEATH RATE† PERCENT OF TOTAL DEATHS 1 Diseases of the heart 194 24.2 2 Malignant neoplasms 186 23.3 3 Chronic lower respiratory diseases 45 5.6 4 Cerebrovascular diseases 42 5.2 5 Accidents (unintentional injuries) 39 4.9 6 Alzheimer disease 27 3.4 7 Diabetes mellitus |
|
|
Term
Three factors (triad of Virchow) |
|
Definition
promote venous thrombosis: (1) venous stasis (e.g., immobility, obesity, prolonged leg dependency [e.g., air travel], age, heart failure [HF]), (2) venous endothelial damage (e.g., trauma, medications), and (3) hypercoagulable states (e.g., inherited disorders, malignancy, pregnancy, oral contraceptives, hormone replacement, hyperhomocysteinemia, antiphospholipid syndrome). |
|
|
Term
Superior Vena Cava Syndrome |
|
Definition
is a progressive occlusion of the superior vena cava (SVC) that leads to venous distention in the upper extremities and head. |
|
|
Term
Isolated systolic hypertension (ISH) |
|
Definition
elevated systolic blood pressure accompanied by normal diastolic blood pressure (less than 90 mmHg). ISH is becoming more prevalent in all age groups and is strongly associated with cardiovascular and cerebrovascular events. |
|
|
Term
Blacks and Cardiovascular |
|
Definition
In blacks, variants of the apolipoprotein L1 (APOL1) gene are associated with hypertension and renal disease.23 |
|
|
Term
Cardiovascular risk factors |
|
Definition
(1) family history of hypertension; (2) advancing age; (3) gender (men younger than 55 and women older than 70 years); (4) black race; (5) high dietary sodium intake; (6) glucose intolerance (diabetes mellitus); (7) cigarette smoking; (8) obesity; (9) heavy alcohol consumption; and (10) low dietary intake of potassium, calcium, and magnesium.19 |
|
|
Term
High levels of angiotensin II |
|
Definition
contribute to endothelial dysfunction, insulin resistance, dyslipidemia, and platelet aggregation and play an important role in the complications associated with the metabolic syndrome |
|
|
Term
|
Definition
Secreted by the adipose cells, to interact with the hypothalamus to control body weight and fat deposition through appetite inhibition and increased metabolic rate. However, chronically high levels of leptin associated with obesity result in resistance to these weightreducing functions and have been found to increase sympathetic nervous system activity, decrease renal sodium excretion, promote inflammation, and stimulate myocyte hypertrophy |
|
|
Term
|
Definition
protein that is produced by adipose tissue but is reduced in obesity. Decreased adiponectin is associated with insulin resistance, decreased endothelial-derived nitric oxide (vasodilator) production, and activation of the sympathetic nervous and renin-angiotensin-aldosterone systems. |
|
|
Term
|
Definition
is caused by an underlying disease process that raises peripheral vascular resistance or cardiac output. If the cause is identified and removed before permanent structural changes occur, blood pressure returns to normal. |
|
|
Term
|
Definition
(rapidly progressive hypertension in which diastolic pressure is usually greater than 140 mmHg) can cause encephalopathy. |
|
|
Term
|
Definition
|
|
Term
|
Definition
(Systolic BP 140-159 mmHg or diastolic BP 90-99 mmHg) Thiazide-type diuretics for most May consider ACE inhibitor, ARB, beta-blocker, CCB, or combination |
|
|
Term
|
Definition
is a localized dilation or outpouching of a vessel wall or cardiac chamber |
|
|
Term
|
Definition
is an extravascular hematoma that communicates with the intravascular space. A common cause of this type of lesion is a leak between a vascular graft and a natural artery.Arteriosclerosis and hypertension are found in more than half of all individuals with aneurysms. |
|
|
Term
loss of medial elastic lamellae and thinning of the tunica media.64 Aneurysms most |
|
Definition
Inflammation (with the production of toxic oxygen radicals) and changes in cytokines, such as TGF-β, activate matrix degrading proteins and smooth muscle cell apoptosis resulting |
|
|
Term
The prevalence of abdominal aortic aneurysms |
|
Definition
2.9 to 4.9 cm in diameter ranges from 1.3% in men 45 to 54 years of age to 12.5% in men 75 to 84 years of age. For women, the prevalence ranges from zero in the youngest to 5.2% in the oldest age group.5 |
|
|
Term
Aneurysms in the heart present with |
|
Definition
dysrhythmias, heart failure, and embolism of clots to the brain or other vital organs. |
|
|
Term
Pulmonary emboli originate |
|
Definition
in the venous circulation (mostly from the deep veins of the legs) or in the right heart |
|
|
Term
Arterial emboli most commonly originate |
|
Definition
in the left heart and are associated with thrombi after MI, valvular disease, left heart failure, endocarditis, and dysrhythmias. |
|
|
Term
Thromboangiitis Obliterans (Buerger Disease) |
|
Definition
is an inflammatory disease of the peripheral arteries. It is associated with smoking in approximately 95% of cases—the other 5% are related to frostbite, trauma, or the use of sympathomimetic drugs.70 |
|
|
Term
There are five primary criteria for the diagnosis of Buerger disease: ( |
|
Definition
1) onset before age 50; (2) history of tobacco use; (3) ischemia of the digits; (4) typical arteriographic findings; and (5) exclusion of autoimmune disease, thrombophilia, diabetes, and proximal embolic source |
|
|
Term
Raynaud phenomenon and Raynaud disease |
|
Definition
is secondary to systemic diseases, such as collagen vascular disease (e.g., scleroderma), chemotherapy, cocaine use, hypothyroidism, pulmonary hypertension, thoracic outlet syndrome, serum sickness, vasculitis, malignancy, or long-term exposure to environmental conditions, such as cold or vibrating machinery in the workplace. |
|
|
Term
|
Definition
is a form of arteriosclerosis in which thickening and hardening of the vessel are caused by the accumulation Once these lipid-laden foam cells accumulate in significant amounts, they form a lesion called a fatty streak (see Figure 32-12). These lesions can be found in the walls of arteries of most of lipid-laden macrophages within the arterial wall, which leads to the formation of a lesion called a plaque. Both innate and adaptive immunity play a role in the development and progression of atherosclerotic lesions.78These macrophages then release enzymes and toxic oxygen radicals that create oxidative stress, oxidize LDL, and further injure the vessel wall. 5. Growth factors also are released, including angiotensin II, fibroblast growth factor, TGF-β, and platelet-derived growth factor, which stimulate smooth muscle cell proliferation in the affected vessel. |
|
|
Term
Peripheral Artery Disease |
|
Definition
refers to atherosclerotic disease of arteries that perfuse the limbs, especially the lower extremities. PAD affects approximately 8 to 12 million Americans, 12% to 20% of whom are 65 years of age or older, and is associated with significant morbidity and mortality. Prevalence increases with age, and PAD disproportionately affects blacks. |
|
|
Term
An increased serum concentration of LDL is a strong indicator of coronary risk. |
|
Definition
LDL is responsible for the delivery of cholesterol to the tissues. Serum levels of LDL are normally controlled by hepatic receptors for LDL that bind LDL and limit liver synthesis of this lipoprotein |
|
|
Term
Low levels of HDL cholesterol also are a strong indicator of coronary risk. |
|
Definition
HDL is responsible for “reverse cholesterol transport,” which returns excess cholesterol from the tissues to the liver, where it binds to hepatic receptors. HDL can remove excess cholesterol from the arterial wall through several pathways, including mediating the efflux of cholesterol from lipid-laden macrophages (foam cells) through the activation of adenosine triphosphate (ATP)–binding cassette transporter proteins (ABC proteins).HDL also participates in endothelial repair and decreases thrombosis. As HDL cholesterol is transported, it progresses through three subtypes of HDL: pre-β HDL, HDL3, and HDL2. Apolipoprotein (ApoA-I) on the pre-β HDL |
|
|
Term
Type I: exogenous hyperlipidemia; fat-induced hypertriglyceridemia |
|
Definition
Cholesterol normal Triglycerides increased three times Chylomicrons increased Abdominal pain Hepatosplenomegaly Skin and retinal lipid deposits Usual onset: childhood Low-fat diet |
|
|
Term
Type IIa: hypercholesterolemia |
|
Definition
Triglycerides normal LDL increased Cholesterol increased Premature vascular disease Xanthomas of tendons and bony prominences Common Onset: all ages Low-saturated-fat and low-cholesterol diet Cholestyraminea Colestipolb Lovastatinc Nicotinic acidd Neomycine Intestinal bypass |
|
|
Term
Type IIb: combined hyperlipidemia; carbohydrate-induced hypertriglyceridemia |
|
Definition
LDL, VLDL increased Cholesterol increased Triglycerides increased Same as IIa Same as IIa; plus carbohydrate restriction Clofibratef Gemfibrozilg |
|
|
Term
Type III: dysbetalipoproteinemia |
|
Definition
IDL or chylomicron remnants increased Cholesterol increased Triglycerides increased Premature vascular disease Xanthomas of tendons and bony prominences Uncommon Onset: adulthood Weight control Low-carbohydrate, low-saturated-fat, and low-cholesterol diet Alcohol restriction Clofibrate Gemfibrozil Lovastatin Nicotinic acid Estrogensh Intestinal bypass |
|
|
Term
Type IV: endogenous hyperlipidemia; carbohydrate-induced hypertriglyceridemia |
|
Definition
Glucose intolerance Hyperuricemia Cholesterol normal or increased VLDL increased Triglycerides increased Premature vascular disease Skin lipid deposits Obesity Hepatomegaly Common onset: adulthood Weight control Low-carbohydrate diet Alcohol restriction Clofibrate Nicotinic acid Intestinal bypas |
|
|
Term
Type V: mixed hyperlipidemia; carbohydrate and fat-induced hypertriglyceridemia |
|
Definition
Glucose intolerance Hyperuricemia Chylomicrons increased VLDL increased LDL increased Cholesterol increased Triglycerides increased three times Abdominal pain Hepatosplenomegaly Skin lipid deposits Retinal lipid deposits Onset: childhood Weight control Low-carbohydrate and low-fat diet Clofibrate Lovastatin Nicotinic acid Progesteronei Intestinal bypass |
|
|
Term
|
Definition
An estimated 47 million residents have a combination of obesity, dyslipidemia, and hypertension |
|
|
Term
Nontraditional Risk Factors. Nontraditional, or novel, risk factors for CAD include: ( |
|
Definition
(1) increased serum markers for inflammation and thrombosis, (2) troponin I, (3) hyperhomocysteinemia, (4) adipokines, (5) infection, (6) air pollution, and (7) coronary artery calcification and carotid wall thickness. |
|
|
Term
Markers of Inflammation and Thrombosis |
|
Definition
Of the numerous markers of inflammation that have been linked to an increase in CAD risk (hs-CRP, fibrinogen, protein C, plasminogen activator inhibitor), the relationship between serum levels of CRP and CAD has been explored in the greatest depth. |
|
|
Term
Other markers of inflammation associated with CAD include erythrocyte sedimentation rate, von Willebrand factor |
|
Definition
von Willebrand factor concentration, uric acid, IL-6, IL-18, TNF-α, fibrinogen, and YKL-40 is a 40 kDa (mass) glycoprotein produced by inflammatory cells, cancer cells, and stem cells |
|
|
Term
Troponin I. Troponin I (TnI) is |
|
Definition
is a serum protein whose measurement is used as a sensitive and specific diagnostic test to help identify myocardial injury during acute coronary syndromes. Highly sensitive TnI assays are used in individuals without a history of CAD to assess risk for future CHD events, mortality, and heart failure |
|
|
Term
|
Definition
Hyperhomocysteinemia occurs because of a genetic lack of the enzyme that breaks down homocysteine (an amino acid) or because of a nutritional deficiency of folate, cobalamin (vitamin B12), or pyridoxine (vitamin B6). Although it has not been identified by the AHA as a major risk factor for CAD, hyperhomocysteinemia is associated with increased LDL oxidation, decreased endogenous vasodilators, increased smooth muscle proliferation, and an increased tendency for thrombosis. |
|
|
Term
|
Definition
Adipokines are a group of hormones released from adipose cells. The two that are most studied are adiponectin and leptin. Leptin is primarily implicated in obesity, hypertension, and diabetes (see |
|
|
Term
|
Definition
Angina pectoris is chest pain caused by myocardial ischemia.The pain is caused by the buildup of lactic acid or abnormal stretching of the ischemic myocardium that irritates myocardial nerve fibers. |
|
|
Term
|
Definition
Prinzmetal angina (also called variant angina) is chest pain attributable to transient ischemia of the myocardium that occurs unpredictably and almost exclusively at rest.Serum markers of inflammation, such as CRP and IL-6, may be elevated in individuals with this form of angina. |
|
|
Term
Nitrates improve coronary blood flow and reduce myocardial |
|
Definition
demand by decreasing peripheral vascular resistance and venous return to the heart (preload) and thereby reduce cardiac workload. Nitrates can be used for short-term symptom relief, and long acting nitrates are available in several forms |
|
|
Term
|
Definition
The process of atherosclerotic plaque progression can be gradual. However, when there is sudden coronary obstruction caused by thrombus formation over a ruptured or ulcerated atherosclerotic plaque, acute coronary syndromes result |
|
|
Term
Myocardial infarction (MI) |
|
Definition
results when prolonged ischemia causes irreversible damage to the heart muscle. |
|
|
Term
|
Definition
Unstable angina is the result of reversible myocardial ischemia and is a harbinger of impending infarction.Unstable angina is a form of acute coronary syndrome that results in reversible myocardial ischemia |
|
|
Term
Diagnosis of unstable angina |
|
Definition
The ECG most commonly reveals ST-segment depression and T-wave inversion during pain that resolves as the pain is relieved.The serum cardiac biomarkers (troponins, creatine phosphokinase-myocardial bound [CPK-MB] and lactate dehydrogenase [LDH1]) remain normal. |
|
|
Term
|
Definition
is acute inflammation of the pericardium. The etiology of acute pericarditis is most often idiopathic or caused by viral infection by coxsackievirus, influenza, hepatitis, measles, mumps, or varicella viruses |
|
|
Term
|
Definition
the accumulation of fluid in the pericardial cavity, can occur in all forms of pericarditis. The fluid may be a transudate, such as the serous effusion that develops with left heart failure, overhydration, or hypoproteinemia.If the fluid is serosanguineous, the underlying cause is likely to be tuberculosis, neoplasm, uremia, or radiatio |
|
|
Term
Restrictive cardiomyopathy |
|
Definition
is characterized by restrictive filling and reduced diastolic volume of either or both ventricles with normal or near-normal systolic function and wall thickness |
|
|
Term
|
Definition
Disorders of the endocardium, the innermost lining of the heart wall, all damage the heart valves, which are made up of endocardial tissue. |
|
|
Term
|
Definition
is the most common valvular abnormality affecting nearly 2% of adults older than 65 years.5 The three common causes are: (1) congenital bicuspid valve, (2) calcific degeneration related to aging, and (3) inflammatory damage caused by rheumatic heart disease |
|
|
Term
|
Definition
Aortic regurgitation results from an inability of the aortic valve leaflets to close properly during diastole resulting from abnormalities of the leaflets or the aortic root, or both. I |
|
|
Term
|
Definition
Mitral stenosis impairs the flow of blood from the left atrium to the left ventricle. Mitral stenosis is most commonly caused by acute rheumatic fever |
|
|
Term
|
Definition
Mitral regurgitation has a variety of causes. The most common are mitral valve prolapse and rheumatic heart disease. Other causes include infective endocarditis, CAD, connective tissue diseases (Marfan syndrome), and congestive cardiomyopathy |
|
|
Term
|
Definition
usually is associated with dilation and failure of the right ventricle secondary to pulmonary hypertension. Rheumatic heart disease and infective endocarditis are less common causes |
|
|
Term
Mitral Valve Prolapse Syndrome. |
|
Definition
a condition in which the anterior and posterior cusps of the mitral valve billow upward (prolapse) into the atrium during systole Mitral valve prolapse is the most common valve disorder in the United States, with a prevalence estimated at 2.4%.5 Mitral valve prolapse tends to be most prevalent in young women. |
|
|
Term
|
Definition
is a systemic, inflammatory disease caused by a delayed immune response to infection by group A betahemolytic streptococci.165 In its acute form rheumatic fever is a febrile illness characterized by inflammation of the joints, skin, nervous system, and heart.166 If untreated, rheumatic fever can cause scarring and deformity of cardiac structures, resulting in rheumatic heart disease (RHD). |
|
|
Term
The classic manifestations of aortic stenosis are angina |
|
Definition
angina, syncope, and heart failure. |
|
|
Term
|
Definition
Aortic regurgitation results from an inability of the aortic valve leaflets to close properly during diastole resulting from abnormalities of the leaflets or the aortic root, or both. It can be congenital (bicuspid valve) or acquired. Acquired aortic regurgitation can be caused by rheumatic heart disease, bacterial endocarditis, syphilis, hypertension, connective tissue disorders (e.g., Marfan syndrome and ankylosing spondylitis), appetite-suppressing medications, trauma, or atherosclerosis |
|
|
Term
Congestive Heart Failure (CHF) |
|
Definition
The heart fails when it is unable to eject blood delivered to it by the venous system.Most common hospital admission diagnosis for persons >65 years old 2. Types a. Left-sided heart failure (LHF; most common type) |
|
|
Term
Right-sided heart failure (RHF) |
|
Definition
Definition—RV cannot effectively pump venous blood into the lungs. • Blood pools under pressure in the venous system (blood builds up behind the failed heart). 2. Pathogenesis a. Increase in RV afterload (increased resistance to blood flow out of the RV) • Examples—LHF (most common cause RHF), PH, PV stenosis, saddle embolus |
|
|
Term
Acute myocardial infarction (AMI) |
|
Definition
Most common cause of death in adults in the United States b. Prominent in males between 40 and 65 years old c. No predominant sex predilection after 65 years old d. At least 25% of AMIs are clinically unrecognizeda. Sequence for developing an AMI (1) An atheromatous plaque is suddenly disrupted (see Fig. 5-15). (2) Subendothelial collagen and thrombogenic necrotic material are exposed. (3) Platelets adhere to the exposed material and eventually form an occlusive platelet thrombus. b. Role of thromboxane A2 in an AMI (refer to Chapter 15) (1) Platelet aggregating agent that contributes to the formation of the platelet thrombus (2) Acts as vasoconstrictor and causes vasospasm of the artery to reduce blood flow |
|
|
Term
|
Definition
Rheumatic heart disease Increased left atrial pressure; left atrial dilatation, atrial fi brillation Increased pulmonary venous pressure, leading to pulmonary hypertension and right ventricular hypertrophy |
|
|
Term
|
Definition
Rheumatic heart disease Left atrial dilatation |
|
|
Term
|
Definition
causes:Senile calcifi cation of normal tricuspid valve Calcifi cation of congenitally bicuspid valve Rheumatic heart disease: Results Left ventricular hypertrophy, due to pressure overload (increased gradient across stenotic valve); ischaemia of hypertrophic ventricular myocardium (angina, arrhythmias, cardiac failure, sudden death) |
|
|
Term
|
Definition
Aortic incompetence Rheumatic heart disease Infective endocarditis Leaking prosthetic valve Aortic root dilatation (aortic dissection, arthritis, Marfan’s syndrome, syphilis |
|
|
Term
Right ventricular failure |
|
Definition
pulmonary embolism • obstructive lung diseases • interstitial lung diseases |
|
|
Term
|
Definition
is a localized dilation of a vessel wall to which the aorta is particularly susceptible. |
|
|
Term
13. The most common sources of arterial thrombotic emboli from the heart |
|
Definition
are mitral and aortic valvular disease and atrial fibrillation. Tissues affected include the lower extremities, the brain, and the heart. |
|
|
Term
|
Definition
is spasm or occlusion of the coronary arteries and is most often the result of atherosclerotic lesions that limit the flow of blood to the heart.results in an imbalance between coronary supply of blood and myocardial demand for oxygen and nutrients such that reversible myocardial ischemia or irreversible infarction may result. |
|
|
Term
|
Definition
assessed by measuring serum enzymes, such as creatinine kinase and troponins, as well as looking for characteristic changes in the ECG. Those individuals at highest risk for complications present with ST-segment elevations on the ECG (STEMI) and require immediate intervention. Smaller subendocardial infarctions are not associated with ST-segment elevations (non- STEMI) but suggest that additional myocardium is still at risk for recurrent ischemia and infarction. Management may include thrombolytic drugs, antithrombotic |
|
|
Term
|
Definition
s a bolus of matter that is circulating in the blood |
|
|
Term
|
Definition
is a blood clot that remains attached to the vessel wall. |
|
|
Term
|
Definition
inflammation caused by a thrombus. |
|
|
Term
Isolated systolic hypertension i |
|
Definition
is an elevated systolic pressure with a normal diastolic pressure. |
|
|
Term
|
Definition
is the result of extremely complicated interactions of genetics and the environment, mediated by a host of neurohumoral effects. |
|
|
Term
|
Definition
s systolic pressure equal to or greater than 160 mm Hg and diastolic pressure equal to or greater than 100 mm Hg. |
|
|
Term
|
Definition
is an abnormal vasospasm of the coronary vessels that causes transient ischemia, unpredictably and usually at rest, and is more common in women. |
|
|
Term
|
Definition
with activity, is often limited, and resolves with rest. |
|
|
Term
|
Definition
s poor myocardial perfusion that does not cause angina and may be associated only with nonspecific symptoms such as fatigue, dyspnea, or a feeling of unease. |
|
|
Term
|
Definition
is typically experienced as transient substernal chest discomfort, ranging from a sensation of heaviness or pressure to moderately severe pain. |
|
|
Term
|
Definition
Approximately two-thirds of Americans older than 60 years of age have hypertension, and approximately 48% of those have adequately controlled hypertension. Hypertension is defined as a diastolic pressure of 90 mm Hg or higher or a systolic pressure of 140 mm Hg or higher. Between 25% and 37% of adults in the United States are prehypertensive. |
|
|
Term
|
Definition
is a yellow lipid ring around the cornea |
|
|
Term
|
Definition
Antibodies are primarily responsible for protection against viruses and bacteria and are part of the humoral arm of the immune system |
|
|
Term
The antigenic determinant or epitope |
|
Definition
s the precise portion of the antigen that is configured for recognition and binding of an antibody. |
|
|
Term
|
Definition
is the matching portion on the antibody or lymphocyte receptor. |
|
|
Term
|
Definition
is one that is not foreign to the host. |
|
|
Term
|
Definition
is an antigen that induces an immune response. |
|
|
Term
The J chain anchors together the IgA molecule |
|
Definition
to form dimers and helps them resist degradation from enzymes. |
|
|
Term
|
Definition
|
|
Term
|
Definition
IgA-2 is found in the secretions of the body. |
|
|
Term
Paroxysmal nocturnal dyspnea (PND) |
|
Definition
occurs when individuals with heart failure or lung disease wake up at night gasping for air and must sit up or stand to relieve the dyspnea. |
|
|
Term
Kussmaul respirations are characterized by |
|
Definition
slightly increased ventilatory rate, very large tidal volume, and no expiratory pause. |
|
|
Term
|
Definition
is commonly caused by disorders such as pulmonary fibrosis that stiffen the lungs or chest wall and decrease compliance. Restricted breathing is characterized by small tidal volumes and rapid ventilatory rate (tachypnea). |
|
|
Term
Labored breathing occurs whenever there is an i |
|
Definition
increased work of breathing, especially if the airways are obstructed, as in chronic obstructive pulmonary disease (COPD). If the large airways are obstructed, a slow ventilatory rate, increased CHAPTER 35 Alterations of Pulmonary Function 1249 1250 UNIT X The Pulmonary System effort, prolonged inspiration or expiration, and stridor (high- pitched sounds made during inspiration) or audible wheezing (whistling sounds on expiration) are typical. In |
|
|
Term
Cheyne-Stokes respirations a |
|
Definition
re characterized by alternat- ing periods of deep and shallow breathing. Apnea lasting 15 to 60 seconds is followed by ventilations that increase in volume until a peak is reached, after which ventilation (tidal volume) decreases again to apnea.Cheyne-Stokes respirations result from any condition that slows the blood flow to the brainstem, which in turn slows impulses sending information to the respi- ratory centers of the brainstem. |
|
|
Term
|
Definition
control of breathing such that minute volume (tidal volume × respiratory rate) is reduced. When alveolar ventilation is normal, carbon dioxide (CO2) is removed from the lungs at the same rate at which it is produced by cellular metabolism. This maintains arterial CO2 pressure (Paco2) at normal levels (40 mmHg). With hypoventilation, CO2 removal does not keep up with CO2 production and Paco2 increases, causing hypercapnia (Paco2 greater than 44 mmHg). |
|
|
Term
|
Definition
s alveolar ventilation that exceeds meta- bolic demands. The lungs remove CO2 at a faster rate than it is produced by cellular metabolism, resulting in decreased Paco2 or hypocapnia (Paco2 less than 36 mmHg). Hypocapnia results in a respiratory alkalosis that also can interfere with tissue func- tion |
|
|
Term
|
Definition
or increased CO2 concentration in the arterial blood (increased Paco2), is caused by hypoventilation of the alveoli.12 |
|
|
Term
|
Definition
(1) depression of the respiratory center by drugs; (2) diseases of the medulla, including infections of the central nervous system or trauma; (3) abnor- malities of the spinal conducting pathways, as in spinal cord disruption or poliomyelitis; (4) diseases of the neuromuscular junction or of the respiratory muscles themselves, as in myasthe- nia gravis or muscular dystrophy; (5) thoracic cage abnormali- ties, as in chest injury or congenital deformity; (6) large airway obstruction, as in tumors or sleep apnea; and (7) increased work of breathing or physiologic dead space, as in emphysema. |
|
|
Term
Acute Respiratory Failure |
|
Definition
Respiratory (lung) failure is defined as inadequate gas exchange, that is, hypoxemia, in which Pao2 is ≤50 mmHg, or hypercapnia, in which Paco2 is ≥50 mmHg with a pH of ≤7.25. |
|
|
Term
|
Definition
Pneumothorax is the presence of air or gas in the pleural space caused by a rupture in the visceral pleura (which surrounds the lungs) or the parietal pleura and chest wall |
|
|
Term
Primary (spontaneous) pneumothorax, |
|
Definition
usually men) between ages 20 and 40 years, is most often caused by the spontaneous rupture of blebs (blister-like formations) on the visceral pleura, although there may be underlying pleural disease with emphysema-like changes.16 Approximately 10% of affected individuals have a sig- nificant family history of primary pneumothorax that has been linked to mutations in the folliculin gene (Birt-Hogg-Dubé- syndrome), which influences cell–cell adhesion.17 Bleb rupture can occur during sleep, rest, or exercise. The ruptured bleb or blebs are usually located in the apexes of the lungs. |
|
|
Term
Secondary (traumatic) pneumothorax c |
|
Definition
an be caused by chest trauma, such as a rib fracture, stab or bullet wounds, or a surgical procedure that tears the pleura; rupture of a bleb or bulla (larger vesicle |
|
|
Term
|
Definition
is most commonly caused by transthoracic needle aspiration |
|
|
Term
|
Definition
is the presence of fluid in the pleural space. The source of the fluid is usually blood vessels or lymphatic vessels lying beneath either pleura, but occasionally an abscess or other lesion may drain into the pleural space. |
|
|
Term
Empyema (infected pleural effusion) |
|
Definition
is the presence of pus in the pleural space. It is thought to develop when the pulmonary lymphatics become blocked, leading to an outpouring of contam- inated lymphatic fluid into the pleural space. Empyema occurs most commonly in older adults and children and usually develops as a complication of pneumonia, surgery, trauma, or bronchial obstruction from a tumor.21 Commonly documented infectious microorganisms include Staphylococcus aureus, Escherichia coli, anaerobic bacteria, and Klebsiella pneumoniaeIndividuals with empyema present clinically with cyanosis, fever, tachycardia (rapid heart rate), cough, and pleural pain. Breath sounds are decreased directly over the empyema |
|
|
Term
Restrictive Lung Disorders |
|
Definition
Restrictive lung disorders are characterized by decreased com- pliance of lung tissue. Pulmonary function testing reveals a decrease in FVC. Restrictive lung diseases can cause ventilation and perfu- sion mismatch and affect the alveolocapillary membrane, which reduces the diffusion of oxygen from the alveoli into the blood and results in hypoxemia |
|
|
Term
|
Definition
is the passage of fluid and solid particles into the lung. |
|
|
Term
There are three types of atelectasis: c |
|
Definition
compression, absorption, and surfactant impairment: 1. Compression atelectasis is caused by the external pres- sure exerted on lung tissue, such as occurs with tumors, or by fluid or air in the pleural space. Atelectasis at the base of the lungs can be caused by abdominal distention pressing on a portion of the lung, causing the alveoli to collapse. 2. Absorption atelectasis results from gradual absorption of air from obstructed or hypoventilated alveoli or from inhalation of concentrated oxygen or anesthetic agents. 3. Surfactant impairment results from decreased produc- tion or inactivation of surfactant, which is necessary to reduce surface tension in the alveoli and thus prevent lung collapse during expiration. Surfactant impairment can occur because of premature birth, acute respiratory distress syndrome, anesthesia, or mechanic |
|
|
Term
|
Definition
Bronchiectasis is persistent abnormal dilation of the bronchi. It usually occurs in conjunction with other respiratory conditions that are associated with chronic bronchial inflammation, such as obstruction of an airway with mucous plugs, atelectasis, aspi- ration of a foreign body, infection, cystic fibrosis, tuberculosis, congenital weakness of the bronchial wall, or impaired defense mechanisms. |
|
|
Term
|
Definition
s diffuse inflammation of the small airways or bronchioles. It is most common in children (see Chapter 36). In adults it usually occurs with chronic bronchitis, but can occur in otherwise healthy individuals in association with an upper or lower airway viral infection (e.g., respiratory syncytial virus [RSV]), or with inhalation of toxic gases. |
|
|
Term
Bronchiolitis obliterans i |
|
Definition
s a late-stage fibrotic process that occludes the airways and causes permanent scarring of the lungs. This process can occur in all causes of bronchiolitis, but is most common after lung transplantation and is associated with acute rejection and infection. |
|
|
Term
|
Definition
is an excessive amount of fibrous or con- nective tissue in the lung. |
|
|
Term
Idiopathic Pulmonary Fibrosis. Idiopathic pulmonary fibrosis (IPF) i |
|
Definition
is the most common idiopathic interstitial lung disorder. It is more common in men than in women and most cases occur after age 60. |
|
|
Term
|
Definition
epresents any change in the lung caused by inhalation of inorganic dust particles, which usually occurs in the workplace. As in all cases of environmentally acquired lung disease, the individual’s history of exposure is important in determining the diagnosis. Pneumoconiosis often occurs after years of exposure to the offending dust with progressive fibrosis of lung tissue. |
|
|
Term
|
Definition
epresents any change in the lung caused by inhalation of inorganic dust particles, which usually occurs in the workplace. As in all cases of environmentally acquired lung disease, the individual’s history of exposure is important in determining the diagnosis. Pneumoconiosis often occurs after years of exposure to the offending dust with progressive fibrosis of lung tissue. |
|
|
Term
Hypersensitivity Pneumonitis. |
|
Definition
Hypersensitivity pneumonitis (extrinsic allergic alveolitis) is an allergic, inflammatory disease of the lungs caused by inhalation of organic particles or fumes. Many allergens (antigens) can cause this disorder, including grains, silage, bird droppings or feathers, wood dust (particularly redwood and maple), cork dust, animal pelts, coffee beans, fish meal, mushroom compost, grain molds, mists from standing water, and fumes from paints and resins. |
|
|
Term
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) |
|
Definition
represents a spectrum of acute lung inflammation and diffuse alveolocapillary injury. In the United States, ALI/ARDS is estimated at 64 to 78 cases per 100,000 people.44 |
|
|
Term
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) r |
|
Definition
The most common predisposing factors for ARDSaregeneticfactors,sepsis,andmultipletrauma(espe- cially when multiple transfusions are received); however, there are many other causes, including pneumonia, burns, aspiration, cardiopulmonary bypass surgery, pancreatitis, drug overdose, smoke or noxious gas inhalation, oxygen toxicity, radiation therapy, and disseminated intravascular coagulation.47,48 |
|
|
Term
Exudative (Inflammatory) Phase (Within 72 Hours). ARD |
|
Definition
he initial lung injury damages the alveolocapillary membrane. Lung injury activates neutrophils, platelets, macrophages, lung epithelial and endothelial cells, and uncontrolled inflammation. Inflammatory mediators include complement, cytokines, arachidonic acid metabolites, platelet-activating factor, reactive oxygen species, and other mediators (specifically tumor necrosis factor [TNF], interleukin-1 [IL-1], and IL-6).49 Activated complement factors and platelet aggregation result in intravascular microthrombus formation and further damage to lung capillaries.50 I |
|
|
Term
ARDProliferative Phase (4 to 21 Days). |
|
Definition
Within 1 to 3 weeks after theinitiallunginjury,thereisresolutionofthepulmonary edema and proliferation of type II pneumocytes, fibroblasts, and myofibroblasts. The intra-alveolar hemorrhagic exudate becomes a cellular granulation tissue appearing as hyaline membranes and there is progressive hypoxemia. |
|
|
Term
Fibrotic Phase (14 to 21 Days). |
|
Definition
after the initial injury, remodeling and fibrosis occur. The fibrosis progressively obliterates the alveoli, respiratory bronchioles, and interstitium, leading to a decrease in functionalresidualcapacity(FRC)andcontinuingV ̇/Q ̇ mismatch with severe right-to-left shunt. The result of this overwhelming inflammatory response by the lungs is acute respiratory failure. |
|
|
Term
The new Berlin definition of ARDS pr |
|
Definition
provides categories of severity based on hypoxemia: mild (200 mmHg < PaO2/FIO2 ≤ 300 mmHg), moderate (100 mmHg < PaO2/FIO2 ≤ 200 mmHg), and severe (PaO2/FIO2 ≤ 100 mm Hg).51a |
|
|
Term
|
Definition
Asthma is a chronic inflammatory disorder of the bronchial mucosa that causes bronchial hyperresponsiveness, constric- tion of the airways, and variable airflow obstruction that is reversible.59 |
|
|
Term
|
Definition
athogenesis of asthma, including those that influ- ence the production of IL-4, IL-5, and IL-13; IgE; eosinophils; mast cells; adrenergic receptors; leukotrienes; nitric oxide; and transmembrane proteins in the endoplasmic reticulum. |
|
|
Term
|
Definition
is the presence of air or gas in the pleural space caused by a rupture in the visceral pleura (which surrounds the lungs) or the parietal pleura and chest wall |
|
|
Term
|
Definition
air pressure in the pleural space equals barometric pressure because air that is drawn into the pleural space during inspiration (through the damaged chest wall and parietal pleura or through the lungs and damaged visceral pleura) is forced back out during expiration |
|
|
Term
|
Definition
however, the site of pleural rupture acts as a one-way valve, permitting air to enter on inspiration, but preventing its escape by closing during expiration. |
|
|
Term
Chronic obstructive pulmonary disease (COPD) |
|
Definition
is defined as a “preventable and treatable disease with some significant extrapulmonary effects that may contribute to the severity in individual patients. Its pulmonary component is characterized by airflow limitation that is not fully reversible. The airflow limitation is usually progressive and associated with an abnormal inflammatory response of the lung to noxious particles or gases.”80 COPD |
|
|
Term
|
Definition
is the third leading cause of death in the United States and is the sixth leading cause of death worldwide.80 Overall mortality from COPD has increased in the United States over the past 30 years; however, mortality in women has increased more than twice that much |
|
|
Term
|
Definition
Pneumonia is infection of the lower respiratory tract caused by bacteria, viruses, fungi, protozoa, or parasites. It is the sixth leading cause of death in the United States and is responsible for more disease and death than any other infection.1 |
|
|
Term
|
Definition
characterized by destruction of alveoli through the breakdown of elastin within the septa by an imbalance between proteases and antiproteases, oxidative stress, and apoptosis of lung structural Cel- lular apoptosis and early cellular senescence contribute to loss of alveolar cells and reduced surface area for gas exchange. Alveolar destruction also produces large air spaces within the lung paren- chyma (bullae) and air spaces adjacent to pleurae (blebs). |
|
|
Term
Pulmonary embolism (PE) i |
|
Definition
s occlusion or partial occlusion of the pulmonary artery or its branches by an embolus. PE most commonly results from embolization of a clot from deep venous thrombosis involving the lower leg |
|
|
Term
Pulmonary artery hypertension (PAH) i |
|
Definition
is defined as a mean pulmonary artery pressure greater than 25 mmHg at rest.159 Pulmonary artery pressure is lower than systemic arterial pressure and is normally 15 to 18 mmHg. |
|
|
Term
|
Definition
Idiopathic PAH is characterized by endo- thelial dysfunction with overproduction of vasoconstrictors (e.g., thromboxane and endothelin) and decreased production of vasodilators (e.g., nitric oxide and prostacyclin).159,160 Release of growth factors causes proliferation of endothelial cells, smooth muscle cells, and fibroblasts, resulting in patchy changes in pre- capillary pulmonary arteries (resistance vessels). T |
|
|
Term
|
Definition
Increased cytosolic calcium (vasocon- striction), phosphodiesterases (modulate cellular proliferation), (vasoconstrictor), and adrenomedullin (vasodila- tor) also play a role in the pathogenesis of this disorder. |
|
|
Term
|
Definition
is secondary to pulmonary artery hypertension and consists of right ventricular enlargement (hypertrophy, dilation, or both).167 |
|
|
Term
|
Definition
Cor pulmonale develops as pulmonary artery hypertension creates chronic pressure overload in the right ventricle similar to that created in the left ventricle by systemic hypertension. |
|
|
Term
|
Definition
(NSCLC, about 85% of all lung cancers) and neuroen- docrine tumors (about 14% of all lung cancers). |
|
|
Term
|
Definition
NSCLC can be subdivided into three common types of lung cancer: squamous cell carcinoma, adenocarcinoma, and large cell undifferentiated carcinoma, and they each have cellular subtypes.1 |
|
|
Term
on–Small Cell Lung Cancer (NSCLC) |
|
Definition
Squamous Cell Carcinoma. Squamous cell carcinoma (SCC) accounts for about 30% of bronchogenic carcino- mas, representing a sharp decline in incidence in the past two decades. |
|
|
Term
Adenocarcinoma. Adenocarcinoma |
|
Definition
tumor arising from glands) of the lung constitutes 35% to 40% of all bronchogenic carcinomas (Figure 35-24, B). Adenocarcinoma occurs more frequently in women, in non-smokers, and in Asians. Envi- ronmental tobacco smoke, occupational carcinogens, viruses, hormones, and positive family history are associated with this tumor type. |
|
|
Term
Neuroendocrine Lung Tumors Small Cell (Oat Cell) Carcinoma. Small cell lung carcinomas (SCLCs) |
|
Definition
are the most common type of neuroendocrine lung tumors and constitute about 14% of bronchogenic carcino- mas, but cause 25% of lung cancer deaths.1 |
|
|
Term
How many genes do humans have |
|
Definition
Humans have approxi- mately 20,000 to 25,000 genes. A |
|
|
Term
Amniocentesis. This procedure, which is usually carried out at |
|
Definition
about 16 weeks' gestation, involves the withdrawal of a small amount of amniotic fluid from the uterus |
|
|
Term
Chorionic villus sampling (CVS). Carried out at |
|
Definition
10 to 12 weeks’ gestation, CVS is performed by extracting a small amount of villous tissue directly from the chorion. |
|
|
Term
Analysis of fetal DNA in maternal circulation. |
|
Definition
y approximately 6 to 8 weeks' gestation, fetal cells can be found in the mother’s bloodstream and these cells (or cell-free fetal DNA) can be tested for some disease- causing mutations. |
|
|
Term
Each DNA subunit—consisting of one |
|
Definition
ne deoxyribose molecule, one phosphate group, and one base (see Figure 4-1)—is called a nucleotide. |
|
|
Term
|
Definition
is the base pair substitution, in which one base pair is replaced by another. |
|
|
Term
|
Definition
This alteration involves the insertion or deletion of one or more base pairs to the DNA molecule. |
|
|
Term
RNA differs from DNA in that |
|
Definition
uracil rather than thymine is one of the four nitrogenous bases. |
|
|
Term
Transcription is the process by which |
|
Definition
RNA is synthesized from a DNA template. The result is the formation of messenger RNA (mRNA) from the base sequence specified by the DNA mol- ecule. Transcription of a gene begins when an enzyme called RNA polymerase binds to a promoter site on the DNA. |
|
|
Term
|
Definition
any of the RNA sequences are removed and the remaining sequences are spliced together to form the func- tional mRNA that will migrate to the cytoplasm. The excised sequences are called introns, |
|
|
Term
|
Definition
The excised sequences are called introns, and the sequences that are left to code for proteins are called |
|
|
Term
Translation is the process by which |
|
Definition
RNA directs the synthesis of a polypeptide |
|
|
Term
The most well-known example of aneuploidy in an auto- some is trisomy of the twenty-first chromosome, which causes |
|
Definition
Trisomy 21 or down's syndrome Down syndrome is seen in 1 in 800 live births |
|
|
Term
|
Definition
|
|
Term
|
Definition
Individuals with at least two X chromosomes and a Y chro- mosome in each cell.1 in 1000 male births. About two thirds of the cases are caused by nondisjunction of the X chromosomes in the mother, and the frequency of the disorder rises with maternal age. |
|
|
Term
|
Definition
Other symptoms include low birth weight, severe mental retardation, microcephaly (smaller than normalhead size), heart defects, and the typical facial appearance shown in Figure 4-17. The disease is caused by a deletion of part of the short arm of chromosome 5. |
|
|
Term
|
Definition
it is associated with substantial cognitive impairment. With a relatively high population prevalence (affecting approximately 1 in 4000 males and 1 in 8000 females), fragile X syndrome is the second most common genetic cause of mental retardation (after Down syndrome). |
|
|
Term
|
Definition
another well-known autosomal domi- nant condition and its main features are progressive dementia and increasingly uncontrollable movements of the limbs |
|
|
Term
The penetrance of a trait i |
|
Definition
is the percentage of individuals with a specific genotype who also exhibit the expected phenotype. Incomplete penetrance means that indi- viduals who have a disease-causing allele may not exhibit the disease phenotype at all, even though the allele and the associ- ated disease may be transmitted to the next generation. |
|
|
Term
variable expressiv- ity in an autosomal dominant disease |
|
Definition
is type 1 neurofibroma- tosis, or von Recklinghausen disease. Like the retinoblastoma gene, the neurofibromatosis gene normally encodes a tumor suppressor.The expression of this gene can vary from a few harmless café-au-lait spots (“coffee with milk,” describing the light brown color) on the skin to malignant tumors, scolio- sis, seizures, gliomas, hypertension, learning disabilities, and neuromas |
|
|
Term
|
Definition
cystic fibrosis, occurs in about 1 in 2500 births. Approximately 1 in 25 whites carries one copy of an allele that can cause cystic fibrosis |
|
|
Term
Important criteria for discerning autosomal recessive inheri- tance include the following: |
|
Definition
1. Males and females are affected in equal proportions. 2. Consanguinity is sometimes present. 3. The disease is seen in siblings but usually not in their parents. 4. On the average, one fourth of the offspring of carrier parents will be affected. |
|
|
Term
|
Definition
(for “sex-determining region on the Y”), |
|
|
Term
Pedigrees for X-linked recessive conditions show the following distinctive features: |
|
Definition
1. The trait is seen much more often in males than in females because females must inherit two copies of the recessive allele (one from each parent) to express the disease, while males need only inherit one copy (from their mother) to express the disease. 2. Because a father can give a son only a Y chromosome, the trait is never transmitted from father to son. 3. The gene can be transmitted through a series of carrier females, causing the appearance of a “skipped generation.”4. The gene is passed from an affected father to all his daugh- ters, who, as phenotypically normal carriers, transmit it to approximately half their sons, who are affected. |
|
|
Term
The most common and severe of all X-linked recessive dis- orders is |
|
Definition
Duchenne muscular dystrophy (DMD), which affects approximately 1 in 3500 males. As its name suggests, this dis- order is characterized by progressive muscle degenerationAffected individuals are usually unable to walk by 10 to 12 years of age. The disease affects the heart and respiratory muscles, and death caused by respiratory or cardiac failure usually occurs before 20 years. |
|
|
Term
The DMD gene is the largest gene ever found in the human, spanning more than 2 million DNA bases. It encodes a |
|
Definition
a previously undiscovered muscle protein, termed dystrophin. |
|
|