You are on page 1of 13

I. CONGENITAL DISORDERS Acute Gastritis Type A (auto-immune) Gastritis Type B (H.

stritis Type A (auto-immune) Gastritis Type B (H. pylori) gastritis: (most common form of gastritis – 90%) –
especially acquired in childhood
Gastroschisis: congenital malformation of the abdominal wall What is it? Acidic damage to the mucosa due to the imbalance What is it? Auto-immune destruction of gastric parietal cells (in
leading to exposure of abdominal contents (so abdominal wall is between acidic environment and mucosal defenses body/fundus of the stomach) What is it? H-pylori induced acute and chronic inflammation of the
split, and abdominal contents are split) – the lateral, anterior, •lined by columnar epithelium stomach (most commonly of the ANTRUM)
posterior… all the walls don’t come together, so you can directly see •Efficient blood supply (if any acid leaks, blood vessel simply takes it How does this destruction happen? Via Type IV HSR (T-cell
the contents of the abdomen (like intestines) à hole in abdominal away) mediated) causes damage in the parietal cells of the stomach How does H. pylori induce inflammation? It produces ureases and
wall •Surface mucus cells/Faviola cells that secrete both: leading to the formation of anti-parietal or anti-IF auto-antibodies proteases that weaken and destroy the stomach’s mucosal defenses (it
• mucus (lubricating + protecting) (they’re consequences) simply sits on the surface of the stomach, does NOT invade)
Omphalocele: persistent HERNIATION of the bowel into the • HCO3 to neutralize any acid that comes close)
umbilical cord due to failure of herniated intestines to go back into What are the clinical features of Type A gastritis? What’s the classical clinical presentation of H. pylori gastritis?
body cavity during fetal development à contents of the abdomen What are common risk factors to developing acute gastritis? 1) As parietal cells knocked out, thickness of mucosa 1) Epigastric abdominal pain
are present in a bubble that is both the AMNION à herniation of •Increase in H+ production/secretion: destroyed à mucosal atrophy 2) Increased risk for:
bowel leading to reveal of contents BUT these abdominal contents • Alcohol-induced (excessive alcohol consumption directly 2) Destruction of parietal cells à Achlorhydria (low acid 1) Ulceration (because chronic inflammation, less
are covered by a bubble-like surface (peritoneum + amnion of damages the mucosa, slightly corrosive) production by stomach, gastrin levels will increase due to defenses, ulcer develops) à peptic ulceration
umbilical cord) • Aspirin (increases H+ secretion) lack of negative feedback, gastrin wants to compensate) 2) Gastric Adenocarcinoma (inflammation leads to
• Cushing Ulcer (increased intracranial pressure will lead to 3) Hyper-gastrinemia + G-cell hyperplasia (due to increased intestinal metaplasia)
Diaphragmatic Hernia: herniation of abdominal contents into the increased parasympathetic/vagal stimulation, which will in turn gastrin secreted to try and increase) 3) MALT lymphoma (inflammation leads to the
thoracic cavity due to weakness or absence of diaphragmatic tissue lead to increased acetylcholine, leading to increased H+ 4) Vit. B12 deficiency + megaloblastic anemia (IF generation of germinal centers within the gastric wall
production.. And indirectly gastrin secretion) destroyed, pernicious anemia à Vit. B12 not absorbed à and formation of post center B-cells in the marginal
Pyloric Stenosis: its an anatomical disorder (+ congenital) •Decrease in mucosal defenses: stops cell division à megaloblastic anemia) zone around the GC à marginal zone leads to MALT
What is it? Hypertrophy (thickening of the wall) of the pyloric smooth • Severe burn (curling ulcer as a result) – leads to hypovolemia, 5) Increased risk for gastric adenocarcinoma (chronic lymphoma, B-cell lymphoma)
muscle more common in MALES (+ infants) à tightening of the less blood flow to stomach (cant sweep away acid) inflammation induces intestinal metaplasia – normally, no
pyloric sphincter so food cannot cross into duodenum • Shock (decreased blood flow to all organs including stomach, less inflammatory cells in the wall of stomach, but in chronic What’s the treatment for H. pylori gastritis? it resolves gastritis and
What is the typical clinical presentation? mucosal protection, leading to multiple ‘stress’ ulcers) inflammation you have a ton of them – so the cells of reverses metaplasia (intestinal)
1) Babies are NORMAL at birth (no pyloric stenosis at birth) – it • NSAID-induced because NSAIDs inhibit prostaglandin production stomach are weirded out and start producing the cells that 1) Triple Therapy
develops about 2 weeks AFTER birth (takes time for stenosis by inhibiting COX, and PGs are essential for: are normally in contact with inflammatory cells like Peyer’s 2) Quadruple Therapy (in case of allergy to clarithromycin)
to develop) 1. Maintaining mucosal barrier (stimulate surface patches = intestinal cells; thus you get intestinal metaplasia)
2) As food goes in stomach, pressure builds up because food mucosal cells to produce mucus and HCO3) How to diagnose H. pylori?
accumulating => PROJECTILE vomiting (not regurgitation) 2. Decrease acid production (act on parietal cells How does Type A gastritis appear on histology? 1) Positive Urea Breath test (so if its negative, you can be sure
that is NON-BILIOUS (because contents haven’t met bile in directly, remember Gi via cAMP) • Foviolar cells (surface mucus cells) wont have big mucus infection is eradicated)
duodenum just yet) 3. Increase blood flow to mucosa barrier (to sweep granules within them 2) Positive Stool antigen (also if negative, confirms eradication)
3) Olive-like mass can be felt on the physical exam – peristalsis away acid) • GOBLET cells that are usually only in the small intestine
is so strong against the sphincter that pressure builds up • Heavy Smoking (intestinal metaplasia) What do we see on histology?
against it leading to hypertrophy (mass) – the actual waves • Radiation and chemotherapy drugs (inhibit regeneration of • Enterochromaffin cell hyperplasia (G-cell hyperplasia) 1) Mucosal chronic inflammation (lympho-plasmocytic infiltrate)
can also be felt gastric mucosa, decrease in defenses) • Lymphoplasmacytic infiltrate within the lamina propria (+ with neutrophils (indicate active inflammation) and lymphoid
What’s the treatment? Myotomy (cut away the muscle) gland infiltration/foci in glands) aggregates
When does it present? A few weeks after birth, more commonly in What are the pathological effects of acute gastritis? (consequences of 2) Intestinal Metaplasia (find intestinal cells in the stomach biopsy
boys + there’s a familial inheritance factor acid damage) Pernicious Anemia (BAB) – a cause of auto-immune gastritis 3) Mucosal atrophy (very important)
1)Superficial Inflammation (Type A) 4) Actual H. pylori bacteria
II. MECHANICAL DISORDERS 2)Erosion (loss of epithelium) These patients lose parietal cells in stomach, thus lose IF à
Mechanical disorders of the stomach can come down to two types: 3)Ulceration (loss of entire mucosal layer) cannot absorb Vit. B12 About H. pylori: it’s a curvilinear gram negative rod that is motile
1) Anatomical (stricture, ulceration…) à Typical findings is high gastrin levels (negative feedback, keep (flagella) and urease positive thus neutralizing the local gastric acid; but
2) Physiological (neural disorders, drug induced) getting secreted in attempt to increase secretion of IF, doesn’t these ureases destroy the gastric mucosa and thus leave the stomach to
Causes of acute gastric ulceration
work) – high gastrin can also lead to glandular hyperplasia be defenseless.
- Shock
III. INFLAMMATION (Gastritis)
- Extensive burns
Acute Gastritis: = burning of the stomach by acid (direct + fast) Virulence = strain dependent (cytotoxic strains are strains with cagA
- Sepsis
that can occur either through: cytotoxin-associated gene or vacA vacuolating cytotoxin)
• Increased acid production - Severe trauma
• Decreased mucosal secretion (protection) - Increased intracranial pressure What are the different types of intestinal metaplasia? The bacteria surrounds its own self with a “bicarbonate cloud” to counter
Type I: complete metaplasia gastric acidity, aided of course by it’s own ureases (it produces urease
Chronic Gastritis: inflammatory changes of mucosa due to acidic à sialomucin expression only which converts urea (abundant in saliva & gastric juices) to
damage eventually leading to ammonia and bicarbonate)
1) Mucosal atrophy Type II: incomplete metaplasia
2) Epithelial Metaplasia (without erosions) à intestinal + gastric type mucin expression What are the stages of gastritis? (Type B)
1) Antral Predominant Stage (inflammation sticks to antrum)
Can be divided into: Type III: incomplete metaplasia 2) Corpus gastritis (inflammation spreads to body and fundus – rest of
• Type A (auto-immune) à involves body/fundus à sulfomucin explession only stomach)
• Type B (H. pylori-induced) à involves antrum 3) Pan-atrophic gastritis (inflammation turns into atrophy of stomach)
• Reflux (bile), chemical gastropathy
Peptic Ulcer Disease Hypertrophic Gastropathies: Benign Neoplasms b. Diffuse Type (expanding, 67%)
1) Menetrier’s disease PRESENTATION:
What is it? Solitary ulcer of the mucosa either involving proximal • WHAT: hyperplasia of superficial mucus cells – so basically you Hyperplastic Polyp (most common gastric polyp – 75-90%) – -- signet ring cells - signet cells are cells with their nucleus pushed to the edge by the
duodenum or distal stomach that breaches the integrity and continuity of would have too much mucus and HCO3 secreted, not enough hyper-proliferation of mucosal (foveolar) epithelium tons of mucin being produced
mucosa and can extend through muscularis mucosa into submucosa or acid in stomach) -- These signet ring cells (tumor cells) diffusely infiltrate the gastric wall (infiltration
even deeper. • CAUSE: excessive production of TGF-alpha (transforming What does it look like? leads to cancer + reactive response of stroma = desmoplasia)
growth factor) • Dome or olive-shaped -- Grow as one mass by expansion – compression of surrounding tissue (diffusely
Why does it form? Due to a disruption of the trilaminar lipoprotein cell • PRESENTATION: epigastric discomfort, diarrhea (no activation • Smooth surface and well-defined margins thickens the wall of the stomach)
membrane of the mucosal cells + pH gradient between cell and of enzymes, maldigestion.. No absorption), weight loss • Majority <1.5 cm RISK FACTORS: (not associated with intestinal metaplasia, H. pylori, or nitrosamines)
lumen of stomach (because you’re not absorbing anything) • Often pedunculated (attached to a long stalk to tissue) HISTOLOGY:
• Solitary (66%) or multiple (<10) -- coherent relationship among cells regardless of maturation/differentiation (not
What are the different types? 1) Hypertrophic-hypersecretory (hyperplasia of parietal cells + individual)
1) Duodenal Ulcer: chief cells – oversecretion of pepsinogen and HCl) Where is it most commonly found? ANTRUM of stomach -- Desmoplasia results in thickening of the stomach wall (linitis plastica)
• CAUSE: H. pylori (about 99% - almost always) and ZE syndrome HEREDITARY DIFFUSE GASTRIC CARCINOMA: inherited, caused by a germ-line
(1% - very rare; production of massive amounts of acid) 2) ZE syndrome (gastrinoma, formation of tumor that keeps on truncating mutation of the CDH1 gene resulting in E-cadherin inactivation leading to
Note: it’s higher in association with certain conditions:
• PRESENTATION: Epigastric pain that improves with meals secreting gastrin leading to continuous acid secretion and pernicious anemia (22-37%), chronic gastritis (6%), partial gastrectomy (4-
loss of cell adhesion BUT increase in cell motility
(duodenum increases defenses and produces neutralizing damage of the mucosal surface + secretory diarrhea) 20%)
substances when anticipates food, thus pain improves because What’s the prognosis?
less acid and corrosion) • WHAT: A gastrin secreting tumor formed due to mucosal à The early gastric carcinoma = confined to mucosa and submucosa (good prognosis
• DIAGNOSIS: endoscopic biopsy showing ulcer + hypertrophy of hyperplasia/hypertrophy (of gastric glands) + excessive acid Adenoma: (7-10% of gastric polyps) – hyper-proliferation of in japan)
BRUNNER glands (mucus producing in duodenal wall) secretion gastric glands (in Japanese people, it’s very common and à Metastasis = worst prognosis
• COMPLICATION: rupture of the ulcer (damage to underlying usually measures <2 cm -- elsewhere in world, >4 cm) -- Immediate metastasis: usually spread to Virchow node (left supraclavicular node)
blood vessel) leading to: • WHERE: In duodenum or pancreas (leftover G cells from fetus) -- Distant metastasis:
• Bleeding from the gastroduodenal artery (posterior What does it look like: soft, velvety, broad based, clear defined 1. Usually involves liver
ulcer) • PRESENTATION: margins, with a flat surface filled with papillary/villous crevices 2. Peri-umbilical region (Sister Mary Joseph nodule) seen with intestinal type
• Acute pancreatitis (posterior ulcer, if activates 1) Abdominal pain that improves with food consumption that go down to it’s base (usually solitary) (infiltrative)
pancreatic enzymes also) (temporarily increases the pH) 3. Bilateral ovaries (Krukenburg tumor) seen with diffuse type (expanding -
2) Gastric Ulcer 2) Chronic Diarrhea due to: (poor digestion, secretory diarrhea, Where is it located: ANTRUM of stomach mucinous)
• CAUSE: H. pylori (70%), NSAIDs (20%), bile reflux (10%) steatorrhea)
• PRESENTATION: Epigastric pain that worsens with meals -- Excess acid secretion denatures pancreatic enzymes Complications: it has the potential to become malignant What’s the presentation?
(because when you start eating, the stomach will secrete more secreted • 6-75% chance to undergo malignant change – especially -- Weight loss
acidic gastric juice and mucosa will be more damaged, even -- Inhibits sodium/water resorption by intestine in larger villous adenomas, not so much small flat -- Abdominal pain
though food temporarily increases pH but not enough) 3) Duodenal ulcers (due to all acid production that washes all adenomas -- Anemia
• DIAGNOSIS: endoscopic biopsy showing ulcer located in the the way down) in the DISTAL duodenum (most occur in the • Can also co-exist with malignant adenocarcinomas (29- -- Early satiety (not hungry) – especially in diffuse type (stomach can’t expand
lesser curvature of the stomach proximal duodenum/duodenal bulb – even jejunum) à 59% of cases) if it’s not malignant itself because it’s so thick)
• COMPLICATION: rupture of the ulcer (damage to underlying refractory to PPI therapy (doesn’t work) -- Acanthosis nigricans – thickening + darkening of skin in axial region
blood vessel) leading to bleeding from the left gastric artery 4) Heartburn (if all that acid gets into the esophagus) -- Leser-Trelat Sign – patient gets dozens of seborrheic keratosis
(posterior ulcer)
• DIAGNOSIS: Malignant Tumors of the Stomach What’s the pathogenesis (how did it develop)? It could have started out as:
What are the causes (differentials) of peptic ulcer disease? H. pylori, 1) Fasting serum gastrin level >10x normal level (fasting to 1. Adenoma
NSAIDs, bile reflux, cancer prevent ‘unwanted’ gastrin secretion triggered by vagal response 2. Chronic gastritis due to H. pylori with associated epithelial changes (most
A- Gastric Carcinoma: the malignant proliferation of surface
- Duodenal ulcers à NEVER malignant (duodenal carcinoma super or stomach distention) epithelial cells (adenocarcinoma) common)
super rare, not really studied/biopsied) 2) Secretin test (differentiates gastrinoma from other causes of
- Gastric ulcers à can be caused by gastric CARCINOMA (thus increased gastrin) à if you give secretin and the gastrin level Can be divided into two types: B- Malignant Lymphoma: the stomach is the most common site of extra-nodal
important to check malignant features) – on endoscopy: increases even more, the ZE syndrome because: a. Intestinal Type (infiltrative, 33%): lymphoma due to the associated lymphoid tissue (MALT lymphoma, mantle cell
- Benign = small, punched out, normal margins around it -- Normally, the G cells inhibited by secretin PRESENTATION: large irregular ulcer with undefined and lymphoma, follicular lymphoma, diffuse B cell lymphoma)
- Malignant = large, irregular, abnormal margins around it -- Gastrinoma are actually stimulated by secretin heaped up margins
What are the possible causes of stomach MALT lymphoma? Genetic changes that
WHERE: lesser curvature of antrum (like in gastric ulcer in
What are the major pathological factors responsible for ulcer • TREATMENT: activate the NF-kappa B pathway
peptic ulcer disease)
development? 1) High dose PPIs (Omeprazole, lansoprazole, pantoprazole) – 1. Fusion of apoptosis inhibitor (API-2) gene and MALT lymphoma associated
RISK FACTORS:
1- Anatomical Factors (disruption of mucosal integrity = worse) standard therapy for ulcers, here try HIGH doses -- Intestinal Metaplasia (chronic gastritis – type A and type B) translocation (MALT1) gene à t(11;18)(q21;q21)
2- Neuroendocrine Status (hormones and neural signals – more vagal 2) Somatostatin (because it inhibits gastrin secretion in addition -- Smoked Foods (nitrosamines) 2. Translocation of BCL10 gene next to immunoglobulin heavy chain gene (IGH) à
stimulation/more gastrin/more acid = worse) to other GI hormones) – also called octreotide -- Blood Type A t(11;14)(p22;q32)
3- Vascular Factors (blood flow to stomach/duodenum – less blood flow 3) Surgical Excision PROGNOSIS: worst
= worse) HISTOLOGY: What is the promoter agent of MALT lymphoma? H. pylori
à Thus cases MALT lymphoma are responsive to H.pylori medication
-- cells how varying degree of differentiation
What are the possible complications of gastric ulcers? Stages of intestinal-type gastric carcinoma are: à Not responsive to antibiotics if:
-- tumor cells are isolated and spread out all throughout tumor
1. Bleeding (15-205) – posterior rupture a- Chronic (active) gastritis -- Specific mutations detected
2. Perforation b- Chronic atrophic gastritis -- Lymphoma deeply infiltrating gastric wall
3. Obstruction c- Intestinal metaplasia -- Transformation to diffuse large B-cell lymphoma (usually diffuse B cell
d- Dysplasia lymphoma that is more aggressive + made up of larger atypical lymphocytes)
e- Carcinoma
Colonic Polyps: are raised protrusions of colonic mucosa 3- Serrated Adenomas: Colonic Adenocarcinoma: (most common malignancy of the colon – Screening: begins at 50 years of age – goal is to (a) remove
96-98% of colorectal cancers, worldwide distribution + Lebanon) adenomatous polyps before carcinoma develops and (b) detect
Types: Cause: combined features of both hyperplastic and adenomatous cancer early. Can be divided into two categories:
1- Hyperplastic: (benign, no malignant potential – BUT right-sided hyperplastic polyps. Most common in elderly females (median age = 61 yrs). 1- Stool tests (fecal occult blood testing, abnormal DNA
polyps are associated with carcinoma) Epidemiology: 3rd most common cancer in world, and 2nd most
common cause of cancer death. testing) à detect cancer (mainly)
Locations: proximal (right) side of colon -- 6% chance of developing colorectal cancer 2- Structural tests (colonoscopy, CT scan, double-contrast
Cause: due to hyperplasia of the glands (most common type of polyp) barium enema) à detect both adenoma + cancer
-- 8% of all malignancies in both men and women
Gross Appearance: >5 mm, Flat surface, sessile (embedded) -- peak incidence is 60-70 years (10% occur <50 yrs)
Location: most commonly on left colon (rectosigmoid) Gross Presentation: obstructing mass (1+) within colon that can
-- equally common in males and females
Microscopically: grow in one of two ways:
Microscopy: -- Inverted L-shaped + serrated crypts (sometimes T-shape) 1- Fungating/raised lesion (usually RIGHT-sided) leading to:
-- Small (<5mm diameter) Location: equal distribution in the four major segments of the colon
-- Base of crypts are dilated (ascending, transverse, descending, sigmoid - mostly, rectum) -- iron deficiency anemia (next to terminal ileum)
-- ‘Serrated’ appearance on surface -- vague pain (not in one quadrant)
-- Narrow base/crypts Genetics: Often have microsatellite-instability mutations and DNA (associated with microsatellite instability pathway)
-- Star-shaped lumen (cross-section) Pathogenesis: (genetic disease by definition) derives from columnar
methylation 2- “Napkin’ circumferential ring around colonic segment
glandular epithelium in colonic mucosa, under influence of:
-- Environmental and dietary habits (low fiber diets, obesity, high (usually LEFT-sided) leading to:
2- Adenomatous: (benign, pre-malignant – may progress to adenocarcinoma via the Types: -- left lower quadrant pain
adenoma-carcinoma sequence) alcohol consumption, low physical activity, smoking)
-- Traditional Serrated Adenomas (TSA) -- Genetic factors (97% sporadic, 3% familial): 2 major molecular -- blood-streaked stool
-- Sessile Serrated Adenomas (SSA) – most common subtype pathways: -- decreased stool caliber (squeezed by ring)
Epidemiology: 4x greater risk if 1st degree family member affected (2nd most 1- Adenoma-Carcinoma Sequence (most common, 80%) (associated with adenoma-carcinoma sequence)
common – increased incidence w/ age, 30% prevalence; increased risk in Blacks) --> like Familial Adenomatous Polyposis
Microscopic appearance of: Staging: CRUCIAL (via CT scan or endoscopic ultrasound)
2- Microsatellite Instability Pathway (microsatellites are repeating (endoscopic ultrasound > CT scan for detecting invaded LN)
Cause: neoplastic proliferation of glands Hyperplastic Polyps: (benign, no cancer risk) T: DEPTH of invasion (+ tumor size)
sequences of non-coding DNA that are replicated during cell division; if
--Small (< 5mm) they are copied the way they are = stability; defect in DNA repair N: Spread to regional lymph nodes
Dysplastic Change: (can show low or high grade) -- Serrated Surface M: Metastasis (distant spread) – especially involves liver
mechanisms = instability) à so other genes are also unstable
-- Loss of Mucin -- Narrow crypts at bottom --> like Lynch Syndrome
-- Nuclear Elongation (hyperchromatic) à cancerous = polymorphic nuclei (different) Stage 0: Carcinoma (CA) in situ
-- Nuclear Stratification (still ordered) à cancerous = haphazardly arranged Serrated Sessile Adenoma: (premalignant) Stage 1: deep CA (superficial to wall), no LN, no metastasis
Presentation: obstructing mass in colon (necrotic. Ulceration,
-- larger (> 5mm) exophytic growth, ‘apple core’ lesion) in addition to: Stage 2: deep CA (through wall), no LN, no metastasis
Cancerous Change: progression into adenoma-carcinoma via sequence: -- Serrated surface -- rectal bleeding (+/- anemia) Stage 3: deep CA, 1-2 LN, no metastasis
1. APC (tumor suppressor gene on chromosome 5 – must knock out both copies) -- Inverted L-shaped Stage 4: v. deep CA, 3-4 LN, metastasis (6 mo-2 yrs)
-- anorexia, weight loss
mutations = increased risk for (adenoma) polyp formation -- Dilated crypts at bottom -- change in bowel function
-- Sporadic = with age, both copies can get knocked out via mutations Treatment: surgical resection + adjuvant/neoadjuvant therapy
(depends on location, see later)
-- Germline = Familial Adenomatous Polyposis Syndrome Traditional Serrated Adenoma: (benign, no dysplasia) (usually 5-FU as chemotherapeutic agent of choice)
2. K-ras mutations = leads to (adenoma) polyp formation -- Tubular/flat surface - Stage I = high cure rate
Metastasis: most common site of distant metastasis is the liver –
3. p53 mutation + increased COX expression = progression to adenocarcinoma -- serrations within glands - Stage II = high relapse rate (5-FU + leucovorin)
diffuse lesions around liver (differential dx is military TB) and:
(after polyp) à aspirin protects against this sequence -- Virchow’s nodes (supraclavicular LN enlargement) - Stage III = 5-year survival ~ 40-45% (5-FU + oxaliplatin)
-- Sister Mary Joseph’s node (presence of umbilical mass) - Stage IV = 5-year survival ~ 6-8%
Gross Appearance: increased risk of cancer with increased size Post-Adenoma Resection Surveillance Intervals -- Ascites (abdominal swelling + fluid accumulation in peritoneum) (radiation therapy used more for rectal cancers)
-- Pedunculated (attached to mucosa via stalk) (when to do colonoscopy)
-- Blumer’s shelf (spread of tumor to pelvis/rectum)
-- Sessile (blends with surrounding mucosa, harder to remove) Notes:
Low-Risk Patients (1-2, small, tubular) ----------------------------- 5 years
High-Risk Patients (2+, large, villous) ------------------------------ 3 years Microscopically:: infiltrative pattern. 1-- patients with colorectal carcinoma have an overall increased
Clinical Manifestations: silent/asymptomatic, with occasional bleeding risk for Streptococcus bovis endocarditis (thus if patient is
V. High Risk Patients (10+, villous) --------------------------------- <3 years -- Infiltrative GLANDS in well to moderately differentiated cases
Post-polypectomy (large adenoma, piecemeal) ----------------- 2-6 months (invade muscle layers, can go into serosa) affected, must perform colonoscopy to check)
Microscopy: can be a range from:
-- Infiltrative CELLS/sheets of cells in poorly differentiated cases
-- Tubular (surface is flat) -- Haphazardly arranged cells 2– an important tumor marker is CEA (elevated in CR cancer,
-- Villous (surface, >80%, covered in finger-like projections) à highest risk of cancer -- mucinous cells (large intracytoplasmic vacuoles containing mucus – but not useful for screening) assesses treatment response
since these are glandular cells, overproduction of mucus) (should decrease) and recurrence.
Screening: very important, can occur via:
1. Colonoscopy (gold standard) since: 3-- chemoprevention of colorectal cancer with NSAIDs (aspirin)
Cytologically: pathologically, colonic adenocarcinoma is graded based
-- gross observation + biopsy testing (to check if hyperplastic or adenomatous polyp) possible as they reduce adenoma formation (and consequent
on the depth of invasion (sometimes, it is so invasive that it’s
-- direct treatment by removing polyps CA development) by inhibiting COX and PG generation – not
penetrates uterus/ovary and get’s mistaken for gynecological cancer)
-- in case if adenoma, what was the risk for the patient to develop carcinoma? celecoxib due to CVA risk.
-- Low-grade (well-differentiated, so the glands are apparent) to high
High risk if: (two most critical factors)
grade nuclear atypia (polymorphic)
(1) Size > 2 cm (advanced adenomas) -- Nuclear overcrowding
(2) Grade of dysplasia – high grade if: -- Increased mitotic activity
-- Sessile pattern (gross)
-- Signet ring cell morphology (indicate more aggressive tumors)
-- Villous surface (histology)
2. Testing for fecal occult blood (only some pathology visible)
Prognosis: dependent on TNM staging and vascular invasion (CT/PET
scan), even before surgery (might have multiple lesions that need to
Treatment: endoscopic resection of adenomatous polyps before progression into
be removed).
carcinoma (invasive) + biopsy/histological evaluation – surgical if endoscopic fails
Other Colonic Neoplasms: II- Mesenchymal Tumors Genetics of Colonic Adenocarcinoma II- Microsatellite Instability – de novo carcinoma

I- Carcinoid Tumor: (low-grade) malignant proliferation of neuro-endocrine A- Gastrointestinal Stromal Tumors (GIST) Mechanism: two distinct mechanisms can cause MMR deficiency:
cells (at bottom of crypts, filled with neuro-endocrine granules, positive for OVERVIEW: (Mechanisms involved) 1- mutations in caretaker genes (MMR genes) -- HNPCC
chromogranin) Pathogenesis: tumor of the interstitial cells of Cajal (pacemaker 2- mutations of BRAF, MAX, IGF2R (MLH1 promoters) – sporadic
Location: occurring anywhere along the gut, but most commonly along the cells of GI tract, found in myenteric plexus, control enteric nervous -- DNA ploidy (aneuploid/diploid)
appendix and small bowel. Appendiceal and rectal carcinoids rarely system) – unlike the other tumors, this one keeps the mucosa intact – -- Mutations (APC, KRAS, BRAF, TP53) Location: right-sided tumors
-- DNA microsatellite instability (MSI)
metastasize. only invades BETWEEN skin layers -- DNA methylation
-- Loss of heterozygosity (LOH) –something abnormal occurs Types:
Pathogenesis: It grows as sub-mucosal polyp-like nodules (raise Location: predominantly in middle aged patients (40-50s): during cell division – once allele is lost entirely and replaced
islands/protrusions, intact mucosa) and tumor secretes serotonin (5-HIAA) -- Small Bowel Tumor (20-30%) 1- Lynch Syndrome (HNPCC, autosomal dominant, mutation of
by a duplication of the other copy/other chromosome.
dumped into portal vein, metabolized in liver by MOA into 5-HIAA, to be -- Gastric Cancer (50-60%) MMR genes – MSH2, MLH1, MSH6, PMS2)
excreted in urine (thus high level of 5-HIAA in urine). No symptoms, yet. -- Colorectal Cancer (<10%) -- Chromosomal loss/gain
Better prognosis than adenocarcinoma. 2- Sporadic (epigenetic silencing like hypermethylation of MLH1 or
Molecular Mechanisms involved in Colon Cancer
Morphological Variants: BRAF mutations) à Hypermethylation of CpG islands (short tandem
1- Genes involved in the replication signaling pathway:
Microscopically: 1- Spindle Cell Type (70%) – elongated nuclei (cigar-shaped) – repeats/promoter sequences) leading to gene silencing = WORSE
-- K-ras (chr. 12)
-- trabeculae/sheets/island of cells (well-defined) nuclei arranged in whorls/fasicles prognosis
-- APC (chr. 5)
-- monotonous (identical) cells with eosinophilic cytoplasm and round 2- Epithelial Type (20%) – round nuclei with abundant cytoplasm -- DCC (chr. 18)
nuclei (high N/C ratio) 3- Mixed (10%) 2- Genes maintaining DNA fidelity (caretaker genes, Clinical Manifestation:
-- minimal pleomorphism + minimal mitotic activity (has to be >2%) -- Early onset
-- positive for neuroendocrine markers like chromogranin (specific, not Immunohistochemistry: microsatellite): MSH2, MLH1, MSH -- more common in females
sensitive) and synaptophysin (sensitive, not specific) – brown color - positive for c-kit (CD117, tyrosine kinase protein) mutation in 80% of 3- DNA methylation (silencing promoter regions) -- associated with serrated polyps (serrated adenomas)
cases à very sensitive, not specific
DNA Repair Mechanisms:
Grading: based on many factors, including: - Some (7/5%) are positive for PDGF receptor alpha Histology: malignant counterpart of SSA
1- Nucleotide Excision Repair
-- Tumor size - DOG1 is a new marker that codes for a Cl- channel protein also -- poorly differentiated + mucinous (signet ring morphology)
2- Base Excision Repair
-- Ki-67 index is an immuno-histochemical stain that looks for proliferative mutated in GIST – more specific than CD117 (which can be found in -- infiltrating lymphocytes within tumor glands
3- Mismatch Repair (MMR) Genes: whose products are
activity (>2% indicates malignancy) other tumors) -- NEGATIVE immunohistochemical stain
critical in detecting and repairing microsatellites (longer
-- Infiltration, angioinvasion, necrosis sequence = more unstable)
Genetics: c-kit is a transmembrane kinase receptor found only on Prognosis:
Carcinoid Syndrome: after the tumor metastasizes into the liver, serotonin mast cells (not neurons) required for development and function of the -- MSI (instable, high-frequency) = better (HNPCC)
interstitial cells of Cajal (ICC). Consequences of Abnormal Repair Genes: -- MSS (stable, low-frequency) = worse (Sporadic)
is dumped into hepatic vein and enters systemic circulation, it then becomes 1- Insertion/Deletion type mutations
-- Normally, c-kit needs to bind to cytokine stem cell factor to be
carcinoid syndrome which is defined by: (only get these symptoms once 2- Chromosomal Instability (CNI) – 85% of Colon Cancer
activated (+ stimulate ICC) Notes:
metastasis to the liver) 3- Microsatellite Instability (MSI) which could be: 15% of
- Bronchospasm -- When mutated, c-kit is continuously activated thus stimulates the Colon Cancer -- Stage II MSI tumors do NOT benefit from 5-FU therapy (harmed)
- Hepatomegaly neoplastic proliferation of ICC (important for targeted therapy) -- we WANT immunohistochemical staining of repair genes to be
-- high frequency (better prognosis, respond more to therapy)
- Diarrhea (intestinal hypermotility) positive (brown)
-- low frequency (difficult to treat, less susceptible)
- Flushing of skin (+ telangiectasia) Staging: we assess the malignant potential of GIST by looking at:
à Release of serotonin from tumor triggered by emotional stress or alcohol 1- Size (>5 cm = malignant) – bigger size, more aggressive
Epigenetic Changes to Genes:
consumption 2- Mitotic Activity (> 5 counts per 50 hypermitotic fields/HPF) Serrated Adenocarcinoma
1- Hypermethylation (silencing) of tumor suppressor genes
B- Smooth Muscle Tumors (Leiomyoma) 2- Hypomethylation (promotion) of proto-oncogenes Epidemiology: 10% of all CRC in females and 6% of males.
Carcinoid Heart Disease: After metastasis, when serotonin gets into the 3- Deamination of methylated cytosine residues leading to
right side of the heart via the vena cava, it causes deposition of collagen (discussed previously – also spindle-like nuclei but negative for c-kit) oncogenic point mutations (C à T) In females: over 18% of adenocarcinomas of the
within the heart valves leading to fibrosis and: ascending colon and cecum.
-- tricuspid regurgitation
-- pulmonary valve stenosis Locations: Cecum (52%, especially in women) and rectum (33%)
Mechanisms of Colorectal Cancers:
(ONLY right sided, because the lung has monoamine oxidase – serotonin III- Malignant Lymphoma (non-Hodgkin, B-cell lymphoma)
gets converted to 5-HIAA before reaching left side of heart) Histology: malignant counterpart of TSA
I- Chromosomal Instability – adenoma-carcinoma sequence
Location: (extranodal) GI tract is the most common site: -- Mucinous differentiation.
Stomach > Small Intestine > Colon > Esophagus -- Eosinophilic cytoplasm.
Mechanism: loss of tumor suppressor genes
-- Vesicular nuclei.
Immunophenotype: most common is MALT lymphoma – other types: -- Absence of dirty necrosis (or ulceration) – main differentiating
Location: left-sided (rectosigmoid) tumors – presents as a
-- Follicular Lymphoma solitary mass factor with SSA
-- Mantle Cell Lymphoma (lymphomatous polyposis – presents as
multiple polyps, aggressive treatment – might need SC transplant) Genetics:
-- Burkitt’s Lymphoma Types: -- BRAF mutation
-- Diffuse large B-cell lymphoma (worst) 1- Sporadic (acquired mutation in APC, p53, DCC, K-ras) -- Microsatellite instability (low-frequency, methylation of MGMT)
2- Familial (FAP, autosomal dominant, APC mutation) -- Microsatellite instability (high frequency, methylation of hMLH1)
3- MUTYH (biallelic, autosomal recessive)
Short Tandem Repeats = unstable non-coding DNA sequences that
Clinical Manifestation: FAP, MUTYH (associated with
are replicated during cell division, thus susceptible to damage
adenomatous polyps)
(especially the CA repeats). In case of any mutation, that will also be
inherited.
CLINICAL
SYNDROME POLYP HISTOLOGY POLYP DISTRIBUTION AGE OF ONSET RISK OF COLON CANCER GENETIC LESION ASSOCIATED LESIONS
MANIFESTATIONS
Familial adenomatous Rectal bleeding, abdominal
Adenoma Large intestine, duodenum 16 yr 100% 5q (APC gene) Desmoids, CHRPE
polyposis pain, bowel obstruction
Possible rectal bleeding,
Orocutaneous melanin
Peutz-Jeghers syndrome Hamartoma Large and small intestine First decade Slightly above average 19p ( STK11) abdominal pain,
pigment spots, other tumors
intussusception
MUTYH-associated Rectal bleeding, abdominal
Adenoma Large intestine, duodenum 45-50 yr 75% (range, 50-100%) 1p ( MYH gene) CHRPE, osteomas
polyposis pain, bowel obstruction
Possible rectal bleeding,
Hamartoma (rarely
Juvenile polyposis Large and small intestine First decade ≈9% PTEN, SMAD4, BMPR1 abdominal pain, Pulmonary AVMs
adenoma)
intussusception
Hereditary nonpolyposis Rectal bleeding, abdominal Other tumors (e.g., ovary,
Adenoma Large intestine 40 yr 30% Mismatch repair genes *
colon cancer pain, bowel obstruction uterus, pancreas, stomach)

I- Familial Adenomatous Polyposis (FAP): autosomal dominant disorder II- Peutz-Jeghers Syndrome – autosomal dominant disorder whereby V- Hereditary Non-Polyposis Colorectal Cancer (HNPCC): (Lynch Syndrome)
characterized by 100s-1000s of adenomatous colonic polyps (if not removed, 100% hamartomatous polyps arise and spread throughout the GI tracts leading to whereby there is a germline mutation leads to an increased risk for colorectal,
will develop cancer). Also called familial polyposis coli (FPC). increased risk for colorectal, breast, and gynecological cancers. ovarian, and endometrial carcinoma.
à Colorectal carcinoma arises from adenoma-carcinoma sequence (usually LEFT à Colorectal carcinoma arises DE NOVO at a relatively early age
sided). Pathobiology: (HIGH penetrance) mutation in DTK11 gene (serine-threonine (usually RIGHT sided).
kinase) on chr. 19 (leading to benign polyp formation)
Genetics: (INCOMPLETE penetrance) due to an inherited APC mutation (tumor Genetics: (HIGH penetrance) due to inherited loss-of-function mutation in DNA
suppressor gene that regulates intestinal epithelial cell growth, on chr. 5) – massive Clinical Manifestations: (average age at diagnosis is in 20s) mismatch repair enzymes (microsatellite instability, found in all cells) – most
risk for development of polyps (1) Mucocutaneous hyperpigmentation in lips, oral mucosa, and genital skin common hereditary colorectal cancer
(2) Small bowel intussusception + obstruction
Clinical Manifestations: Adenomas begin in teen years, and clinical Sx in 20+ years, (3) GI bleeding Clinical Manifestations: (median age for HNPCC is in the mid-40s) – increased
which can have one of three variants (all include FAP -colon polyps, increased risk risk for intestinal (colonic, gastric, pancreatic) and extra-intestinal cancers
of adenocarcinoma): (especially ovarian, endometrial), skin lesions (Tuir-Morre variant)
III- MUTYH-associated polyposis – autosomal recessive disorder characterized
-- Gardner Syndrome: FAP and: by colonic polyposis and high rate of colorectal cancer. Diagnosis: clinically, defined by the presence of all three:
1- Mesenteric fibromatosis (non-neoplastic proliferations in retroperitoneum) (1) Colorectal cancer involving at least 2 generations
2- Osteomas (benign neoplasms most commonly in skull - mandibular) Genetics: mutation leads to defects in base-excision repair (+ other acquired (2) 1+ family member with colorectal cancer <50 yrs
3- Congenital Hypertrophy of retinal pigment epithelium (CHRPE) mutations in APC gene, K-ras gene… lead to CA via adenoma-carcinoma (3) 3+ family members with HNPCC (histologically verified, by presence of
4- Soft tissue tumors (sebaceous cysts, desmoid tumors) sequence) cancer) – at least 1 is 1st degree relative of others + FAP negative
5- Super-numerary teeth

-- Turcot Syndrome: FAP and brain/CNS tumors (medulloblastoma tumor, glial IV- Juvenile Polyp – non-neoplastic tumor made up of tissue normally present but
tumor) disorganized (hamartomatous, benign) usually presents as a solitary rectal polyp
that prolapses and bleeds (can lead to anemia).
-- Attenuated FAP with less than 100 colorectal polyps
Juvenile Polyposis – a whole bunch of juvenile polyps in stomach and colon –
Treatment: prophylactic removal of colon + rectum (or else all patients will develop since large number, increased risk of adenocarcinoma.
carcinoma by 40 yrs)
Familial Adenomatous Polyposis Lynch Syndrome
Small Intestine (Bowel) Types of Mechanical Obstruction Ischemic Bowel Disease: (infarction, ischemic enteritis)
(usually all treated by surgical resection)
Congenital Disorders Causes: – elderly patient with a/ fib, diabetes, vasculitis…
1) Volvulus: twisting of the bowel along its mesentery (180 degrees or 1- Transmural Infarction (thrombosis or embolism of SMA or
1) Duodenal Atresia: (ends in a blind loop) congenital failure of the small more) leading to obstruction of bowel and disruption of blood supply to thrombosis of mesenteric vein)
bowel to turn into a functional duct – usually associated with Down syndrome that part of mesentery (infarction) due to resulting pressure. Surgical 2- Mucosal Infarction (slight decrease of blood supply due to marked
resection only treatment. hypotension)
Clinical Features:
-- Polyhydraminios (too much amniotic fluid) Most common locations are locations with a ton of mesentery: Where: in watershed zones of GI tract (where arterial supply end, no
-- distention of stomach and blind loop of duodenum (“double-bubble sign”) -- sigmoid colon (elderly) direct major blood supply, most susceptible to ischemic injury) like
-- bilious vomiting -- cecum (young adults) splenic flexure of colon, and watershed area of SMA – less
commonly in the sigmoid colon/rectum where IMA/pudendal/iliac
Why polyhydramnios? Remember that amniotic fluid is mostly generated form 2) Intussusception: invagination/telescoping of the proximal segment artery circulations end
the urine of the baby – and at the same time, we resorb some of that fluid by of the bowel (intussusceptum) into the distal segment
swallowing that fluid (thus decreasing the volume). Without that physiologic (intussuscipiens), in the direction of peristalsis. Clinical Features: hypoxic injury followed by reperfusion injury
swallowing/resorption, too much amniotic fluid results. 1- Abdominal Pain
Associated with: 2- Bloody Diarrhea
Why double bubble sign? Both the stomach and the duodenum would be -- infarction (+leading edge) 3- Decreased Bowel Sounds
distended, but the pyloric sphincter is very tight so it appears as two bubbles. -- currant jelly stools (consider it if physical exam not representative of clinical symptoms –
tests negative, but patient in excruciating pain)
Why bilious vomiting? because bile released into the duodenum with nowhere Most common cause: (that creates the leading edge)
to empty, goes back up -- lymphoid hyperplasia after viral infection in the MALT whereby Histology: washing away of mucosal lining (no inflammatory infiltrate,
terminal ileum invaginates into cecum (children) except if superimposed bacterial infection)
2) Meckel’s Diverticulum: outpouching of all three layers of bowel wall -- tumor acts as the lading edge (adults)
(TRUE) due to a failure of the vitelline duct (omphalomesenteric) to Note:
completely involute. 3) Adhesions: fibrous tissue developing after surgical procedures, -- the small bowel needs a lot of ATP due to ton of digestion and
infection, or endometriosis. Leads to abdominal pain, which can be absorption necessary, thus highly susceptible to ischemic injury.
What is the vitelline duct? Early in life, the midgut receives its nutrients from episodic OR requires more surgery (vicious cycle). -- vasculitis or a. fib would cause thrombosis/embolism of SMA;
the yolk sac via the vitelline duct (vital to receiving nutrients). It forms at 4th polycythemia vera or lupus anticoagulant (anti-phospholipid
week, and involutes (resorbs/closes) at 7th week. Persistence leads to 4) Adynamic (paralytic) ileus: paralysis of a portion or of all the syndrome) would cause thrombosis of mesenteric veins
Meckel’s diverticulum. If the duct does not involute at all, there will be passing intestinal tract due to acute intestinal obstruction in the absence of
of muconium via umbilicus – vitelline fistula. mechanical/organic obstruction – halt of bowel movements. Surgical
resection in very advanced cases. Could be due to drugs, Irritable Bowel Syndrome (IBS): relapsing abdominal pain with
Clinically, if you touch these patients you will fell a hardening at umbilicus neuromuscular disorders. bloating, flatulence, and change in bowel habits (either diarrhea and
area (as if there are stools within; solid but soft). That’s because stool will be constipation) that improves with defecation. Classically seen in
caught in Meckel’s diverticulum. middle-aged females.
Types of Hernias:
Features: (rule of 2s) Cause: related to disturbed intestinal motility, but NO identifiable
-- seen in 2% of the population (most common congenital anomaly of GI tract) Inguinal (direct/medial or indirect/lateral – depending on position pathologic changes.
-- it is 2 inches long located in the small bowel within 2 feet of the ileocecal relative to inferior epigastric vessels), Umbilical, Femoral, or Scrotal –
valve due to defects in the muscular wall (abdominal). Treatment: increased dietary fiber may improve Sx
-- presents within first 2 years of life
Incarcerated: hernia CANNOT be reduced (trapped), but there is still a
Clinical Presentation: most cases are asymptomatic, but those that do bit of blood flow to organ. Have a bit more time to plan surgery
present with: (SURGICAL resection is only treatment)
-- perforation/bleeding (because there is ectopic/heterotopic gastric Strangulated: hernia CANNOT be reduced, and there is loss of blood
mucosa and other abnormal tissues within the diverticulum, which can flow to herniated segment leading to ischemic injury with tissue
produce acid and destroy the normal mucosa, leading to bleeding) à may necrosis. Could be life-threatening – emergency.
lead to septic shock if bacteria leaks through
-- intussusception (if large enough)
-- obstruction
-- ischemic damage (if large enough and strangulates bowel itself)
-- volvulus (obstruction caused by twisting)
-- other symptoms depending on what ectopic tissue harbored
Symptoms mimicking appendicitis (inflammation) due to it’s location on the
RIGHT side

3) Duplication: separate loops of intestine share a common blood supply


with a major loop.

4) Enteric Cysts: thin-walled cystic formations in GI tract. The danger is that


it might cause obstruction.
Large Intestine (Colon) C- Vascular Disorders (3) Viruses: most common cause of infectious colitis (esp. in children)

A- Hirschprung disease: (congenital) defective relaxation and peristalsis 1) Angiodysplasia: (acquired) malformation of mucosal and submucosal Pathogens:
of the rectum and distal sigmoid colon – associated with Down Syndrome. capillary beds that arises in the cecum and right colon due to high wall -- rotaviruses (Responsible for destruction of enterocytes in small
tension. More common in older adults, present also as hematochezia. intestine)
More common in boys (1 in 5000 live births – not too rare). -- calciviruses (Norwalk-like agent, in seafood, most common non-
THUS – hematochezia can be caused by: bacterial agent causing food poisoning/gastroenteritis in adults)
Pathophysiology: failure of ganglion cells to descend (from neural crest) High stress (left colon) à Diverticuli -- adenovirus
into submucosa (submucosal plexus, regulate blood flow and secretion) High stress (right colon) à Angiodysplasia (+ mainly elderly) -- astrovirus.
and muscular layers (myenteric plexus, needed for peristalsis and High stress (perianal area) à Hemorrhoids
relaxation) of the large intestine (absence of ganglion cells in segments). Clinical Manifestation: Short incubation period followed by vomiting
2) Hemorrhoids (St. Fiacre’s curse): is the variceal dilatation of the anal and watery diarrhea.
Genetics: at least eight susceptibility genes that control the migration and and peri-anal venous plexus, most commonly associated with
differentiation of ganglion cell precursors into intestinal walls from neural constipation (increased intraluminal venous pressure). Could be internal Pathophysiology: activation of the enteric nervous system (increased
crest cells are associated, including: or external. Especially seen in women due to venous stasis secondary to intestinal motility) leading to diarrhea.
-- RET gene pregnant/gravid uterus in the pelvis, pushing down on the veins.
-- GDNF gene (glial-derived neurotrophic factor) Treated medically or surgically. Histopathology: Shortened villi and lymphocytic infiltration
-- Endothelin gene
-- endothelin receptor gene (ETR3/ETRB) 3) Hereditary Hemorrhagic Telangiectasia: autosomal dominant (4) Opportunistic Infections: (intestinal) in patients with immune
(genes show carriable penetrance) disorder resulting in dilated thin-walled blood vessels, especially in the deficiency states (up to 60% of HIV infected subjects have diarrhea)
nasopharynx and GI tract. Rupture of these blood vessels presents as • Giardia (Giardia lamblia)
Clinical Features: (due to lack of peristalsis/relaxation + blood bleeding (spots). • Crytosporidia (Cryptosporidium parvum) – sits at edges of cells
flow/secretion regulation) • Microsporidia (Enterocytozoon bieneusi, Septata intestinalis,
-- failure to pass meconium (dark green substance, first feces of infant) D- Inflammatory Disorders Encephalitozoon cumiculi).
-- empty rectal vault on digital rectal examination (DRE) • Cytomegalovirus, herpesvirus
-- massive dilation of bowel proximal to obstruction with risk for rupture, 1) Radiation Enteritis: damaging effect of radiation therapy on the • Mycobacterium avium-intracellulare – AIDS-defining
looks like malnourishment mucosa of the GI tract, leading to vascular damage and ischemic injury.
-- constipation (if continued) 4) Pseudomembranous Colitis (antibiotic induced): characterized by
Sometimes, muscle hyperplasia occurs to compensate for loss; makes 2) Ischemic Colitis: (inflammatory/necrotizing) ischemic damage to the diffuse punctate formation of adherent inflammatory exudates
things even worse. colon leading to acute necrotizing inflammation spreading to both small (pseudomembrane – a cap of bacteria cells) overlying injured mucosa.
and large intestine. Most common acquired GI emergency of neonates Seen mostly following broad spectrum antibiotic therapy (C. difficile
Diagnosis: lack of ganglion cells on rectal suction biopsy (normal biopsy (especially premies) – immediate surgery overtakes intestinal flora)
only takes mucosa)
Pathogenesis: atherosclerosis of SMA blocking blood flow to 5) NSAID-induced Colitis: has both acute and chronic manifestations
Treatment: resection of the area of involved bowel (keep the bowel w/ bowel/colon.
ganglion cells proximal to diseased segment)
Affected Sites: splenic flexure, terminal ileum, ascending colon (right
Note: what does a ganglion cell normally look like? Single nucleus, side)
abundant cytoplasm embedded in the thick muscle layer.
Clinical Presentation:
B- Colonic Diverticuli: (false/pseudo-diverticulum) outpouching/herniation -- weight loss (pain, don’t eat as much)
of both mucosa and submucosa through muscularis propria – especially in -- post-prandial pain (after eating, when energy requirement of gut
the left colon. Increased incidence with age (seen in older adults). increases)
-- bloody diarrhea (in case of complete infarction, continued/severe lack
Location: sigmoid/left colon most common (especially in areas where of oxygen)
vasa recta crosses muscularis propria – forms a weak point in the colonic
wall – diverticulum thinning it out) – most common diverticulum of GI tract 3) Infectious Colitis: could be caused by a variety of agents:

Cause: (related to wall stress/increased intraluminal pressure, anything (1) Bacteria: more common in immunocompromised patients
that increases wall stress will push the layers out)
-- Constipation Pathogens: V. cholera, Salmonella, Shigella, Whipple’s disease
-- low fiber diet (Tropheryma whipplei), Mycobacterium tuberculosis

Complications: usually asymptomatic, but complications include: Pathophysiology:


-- Obstruction (with fecal material) – rock-hard mass, looks like a tumor (1) Ingestion of preformed toxin (S. aureus, C. perfringens)
-- Inflammation (diverticulitis) à left-sided appendicitis-like symptoms (2) Infection by toxigenic organism that release toxins (E. coli, V.
-- Hematochezia (rectal bleeding/perforation because diverticulum right cholera, C. jejuni)
next to vein/vasa recta) (3) Infection by enteroinvasion (Shigella, Salmonella, Yersinia
-- Colo-vesicular Fistula (when wall inflamed and ruptures, it bursts and enterocolitis, E. coli, C. jejuni
attaches to a nearby tube like the bladder via adhesions) à presents as
air/stool in urine (2) Protozoa: Entamoeba histolytica, Giardia, nematodes (roundworms),
flatworms.
Note: Diverticulosis (still intact), Diverticulitis (perforated/inflamed)
Diarrhea Malabsorption: (impaired absorption of nutrients) divided into:
Secretory Diarrhea Osmotic Diarrhea IV- Chronic Diarrhea: could be caused by:
I- Definition (most important) Primary Malabsorption – from congenital defects in membrane
There are three ways to define diarrhea – one satisfied criteria is enough Volume > 1 L/day < 1 L/day Irritable Bowel Syndrome (IBS) transport system of small intestinal epithelium
1) FREQUENCY: > 3 bowel movements per day (less than 3 per
week is constipation) Timing Night + Day Day Functional Diarrhea (classified differently than IBS) Secondary Malabsorption – caused by:
2) CONSISTENCY: Abnormal stool consistency (v. important) Pts should have continuous or recurrent passage of loose (mushy) 1) Defects in the epithelial ABSORBTIVE surface
Fasting Effect Diarrhea continues Diarrhea stops or watery stools in >75% of stools WITHOUT abdominal pain or
(Watery, soupy, viscous, semi-formed…) 2) Maldigestion (won’t absorb it if not in correct form)
3) WEIGHT of the stool > 200-300 gms/d (best objective way, not discomfort for at least 3 months with symptom onset at least 6
really practical) – shows you the importance of the IC valve To differentiate between secretory and water diarrhea: months before diagnosis MANIFESTATION: pale, greasy, voluminous, malodorous stools w/
• Weight Loss (calories malabsorbed)
Diarrhea could also be defined based on chronicity: 1) Measure stool electrolytes Chronic Infections (persisting infections like Giardia or C. difficile – • Steatorrhea (fat malabsorbed)
-- ACUTE diarrhea – lasts for < 14 days à mostly due to infectious especially in patients with known risk factors like an • Edema (proteins malabsorbed)
causes (gastroenteritis), could be a bit prolonged in case of alteration of 2) Calculate osmotic stool gap – it’s a calculation that takes into immunosuppressed state, consumption of contaminated water…) • Bloating (lactose malabsorbed)
flora (but still less than a month!) account the concentration of Na+ and K+ ions in the stool.
-- PERSISTENT diarrhea – lasts for > 14 days Cholecystectomy (self-resolving) caused by EXCESSIVE bile acids (1) Lactose Intolerance: (congenital or acquired) due to decreased
-- CHRONIC diarrhea – lasts for > 28 days (VERY important – we start to Normally: Osmotic Gap = 290 – 2([Na+] + [K+]) – b/w 50 & 250 entering the colon (no control b/c no gallbladder). This overwhelms function of lactase enzyme in the brush border of enterocytes,
think about IBS, IBD…) à blood is HYPER-osmotic relative to stool (less electrolytes, more the capacity of the terminal ileum to reabsorb bile salts; the presenting with abdominal distention and diarrhea upon consumption of
water/diluted) remaining bile salts are either: milk products.
II- Major Pathologic Mechanisms: disruption of normal physiology -- taken up by colon (5%)
1) Decreased ABSORPTION of fluid and electrolytes Secretory: <50 mOsm/kg -- stays in colon and pulls water (cholerheic diarrhea) Pathophysiology: (congenital or with age) crush border lactase is lost,
2) Increased SECRETION of fluid and electrolytes à barely any osmotic gap between blood and stool (since a LOT of -- overwhelms porto-hepatic circulation when reabsorbed (thus, to thus lactose cannot be digested. Since (unfermented) lactose is
3) Increased luminal OSMOLALITY (release more osmotically active water is being released continuously without osmotically activate compensate, liver stops water absorption and promotes release of osmotically active, will draw water towards itself.
molecules, so more water has to be released also) molecules) electrolytes – worsens diarrhea)
4) Changes/increased gut MOTILITY à Treated with bile acid sequestrant Clinical Presentation: watery diarrhea (osmotic), abdominal distention
Osmotic: >125 mOsm/kg and bloating (CO2 and H2 gases produced by colonic bacteria).
III- Mechanistic Classification à greater osmotic gap between blood and stool because many Inflammatory Bowel Disease
osmotically active molecules (lactose, sorbitol) are pulling out the water Treatment: empiric lactose-free diet (leads to resolution of symtpms)
Water Diarrhea (any lesion in GI tract – bowel/colon) Microscopic Colitis –
Testing: if necessary, Hydrogen Breath Test (after oral ingestion of
(1) Osmotic = secretion during day only Inflammatory Diarrhea (usually indicates colonic origin) Diarrhea: CHRONIC (intermittent) + WATERY (secretory) lactose to check if there is overproduction due to lactose fermentation
diarrhea w/out bleeding –up to 2L/day of diarrhea EVERY DAY by gut microflora)
Causes: osmotically active agent pulling out water, like Diarrhea: filled with blood and mucus (could start out as watery, without
-- Sorbitol (sugar substitute) Epidemiology: Usually in middle-aged females, but kids can have
blood; if watery doesn’t 100% rule out inflammatory) – small volume
-- Lactose (diagnosed via hydrogen breath test or clinically) it too. (2) Fat Malabsorption (95% of ingested lipid absorbed normally)
-- Osmotic Laxative (lactulose, magnesium citrate) Presentation: frequent bowel movements, tenesmus, fever, severe
abdominal pain (systemic) - NO dehydration Types: Can be divided into two subtypes: Causes: could be due to: among other things
Diarrhea: usually after consumption of osmotic agent 1. Lymphocytic Colitis (you see epithelial lymphocytes on biopsy) A- Chronic pancreatitis (pancreatic insufficiency, no digestion of fat)
Testing: 2. Collagenous Colitis (NO lymphocytic infiltrate) B- Whipple’s disease (mucosal damage of small bowel)
Further Tests: if osmotic agent identified, just remove it from diet (no 1. Fecal calprotectin (magic marker) - contained in cytoplasm of C- ZE syndrome (inactivation of pancreatic lipase by acid, less lipolysis)
further work-up needed) neutrophils, usually shed in stool à when it goes above the cut-off point, Diagnosis: Colonic biopsy (histologically) from colonoscopy. D- Cystic Fibrosis
Macroscopically (observation), the colonic mucosa looks normal –
it signals inflammatory issues present + endoscopy needed
(2) Secretory = continuous secretion (night/day) 2. Endoscopy (to show abnormal gut morphology) maybe with a bit of edema/erythema. Clinical Presentation: steatorrhea
3. Occult Blood (will find fecal leukocytes)
Causes: Testing: Stool test for fat while patient is on 100 gm fat diet; of positive
-- Endocrine (imbalance in gut hormones) Differential Diagnosis: Inflammatory Bowel Disease (UC/CD) then further testing needed:
-- Tumor (secreting fluid/electrolytes) -- pancreatic imaging (check for pancreatitis)
Note: -- secretin stimulation test (gold standard for pancreas function)
Diarrhea: LARGE amounts of fluid shed CONTINOUSLY (>1 L/day) Fatty Diarrhea (known as steatorrhea) -- small bowel biopsy (check for Whipple’s)
Organic disease is the term used to describe any health condition in
which there is an observable and measurable disease process
Effect of Fasting: unaltered – continued (because pathological cause is Diarrhea: greasy, malodorous à If diarrhea accompanied by weight loss + waking up at night to go
still there; like continuous gastrin secretion in ZE syndrome) (3) Tropical Sprue: damage to the small bowel villi due an unknown
to bathroom, indicates organicity
Differential Diagnosis: organism resulting in malabsorption and diarrhea.
Further Tests: -- Fat malabsorption (Especially if accompanied by weight loss) Very similar to celiac disease, except:
Functional disease is when you do all these lab tests and 1- occurs in Tropical regions (like Caribbean) where infectious agent is
1. Stool cultures (to rule out chronic infection) -- chronic pancreatitis everything and they’re normal but the patient is not and is still having located
2. Imaging of small and large bowel (to check for any tumors or
symptoms, such as: 2- occurs after infectious diarrhea (thus, responds to antibiotics)
polyps, or changes in mucosa) Testing: quantitative stool fat measurement (GOLD standard) – need to • Irritable Bowel Syndrome (IBS) – normal blood tests and 3- damage mainly to jejunum and ileum , duodenum is less involved.
3. Selective testing for secretory molecules like: consume >100 g of fat before study; positive result is 7 g of fat in stool calprotectin, BUT
1. Gastin (useful to rule out ZE syndrome or Gastrinoma) per 24 hours • Chronic fatigue syndrome (CFS)
à symptoms tie back to what gets absorbed in the jejunum (folic acid)
2. Vasoactive Intestinal Pilypeptide (to rule out VIPoma) and ileum (Vit. 12).
• Fibromyalgia
3. Serum serotonin or urine 5HIAA (to rule carcinoid)
(4) Celiac Disease: it’s an immune-mediated damage of the small (5) Whipple Disease: (inflammatory colitis) systemic tissue damage to
bowel villi due to gluten exposure associated with HLA DQ2/DQ8. (all around body) characterized by:
-- Histology shows foamy macrophages (sheets of histiocytes) that stain
Genetics: associated with HLA DQ2 and DQ8 (MHC-II) and sometimes positive for PAS
with HLA B8 (MHC-I) -- macrophages’ lysosomes are filled with partially destroyed Tropheryma
Activation of IL-5 by these peptide sequences also mediates the whippelii organisms
immune response. -- macrophages pile up in the lamina propria of small bowel and
compress the lacteals
Pathophysiology: type IV hypersensitivity to gluten (alcohol soluble -- results in fat malabsorption and steatorrhea (chylomicrons can come
protein found in wheat, barley, and rye through)
1- gluten is broken down into gliadin (most immunogenic component, -- common sites of involvement include:
rich in Pro residues, which prevents digestion by enteric enzymes) * small bowel (main)
2- gliadin deamidated by tissue transglutaminase (tTG) * synovium of joints (arthritis)
3- deamidated gliadin taken up and presented on MHC-II by APCs * cardiac valves
4- Helper T cells mediate tissue damage * lymph nodes
à all nutrients are malabsorbed due to villous atrophy * CNS

Histopathology: (6) Abetalipoproteinemia: autosomal recessive deficiency of


-- diffuse villous atrophy (+ blunting of villi – much shorter) apolipoprotein B-48 and B-100
-- loss of circular mucosal duodenal folds, fissures, grooves -- B-48 (carries chylomicron + contributes to formation) à fat
-- basal crypt hyperplasia malabsorption
-- intraepithelial lymphocytosis (lymphocytes in epithelium itself) -- B-100 (carries VLDL and LDL in plasma) à absent plasma VLDL and
-- inflammatory cell (lymphocytes) infiltration of lamina propria LDL

Clinical Presentation:
(7) Small Intestinal Bacterial Overgrowth (SIBO): when endogenous
Children: malabsorption defenses against bacterial overgrowth impaired.
-- abdominal distention/bloating
-- diarrhea Normal Endogenous Defenses:
-- failure to thrive (FTT, underweight, short stature) -- Gastric Acid (#1 barrier)
-- intestinal motility (stasis – bacterial proliferation)
Adults (20s-30s) -- Intact ileocecal valve
-- iron deficiency anemia (celiac work-up = must, villi destroyed) -- immunoglobulins in intestinal secretions
-- megaloblastic anemia (Vit B12 deficiency; villi destroyed) -- bacteriostatic properties of pancreatic/biliary secretions
-- chronic diarrhea (malabsorption)
-- abdominal distention/bloating Etiology: (cases)
-- osteoporosis (due to calcium deficiency; check bone densitometry) -- disorders interfere w/ defenses (achlorhydria, pancreatic insufficiency)
-- glossitis, with recurrent ulcers -- anatomical causes (fistula, diverticula, ileocecal resection)
-- dermatitis herpetiformis (exclusive to CD; due to deposition of IgA -- Motility issues (scleroderma, obstruction)
at tips of dermal papillae, leading to destruction of DE junction, with -- IBS, Crohn’s disease, short bowel syndrome
formation of blisters and vesicles at dermal papillae)
-- increased risk of lymphoma (enteropathy-associated T-cell Location: duodenum and proximal ileum contain small numbers of
lymphoma) and small bowel carcinoma, despite good dietary control bacteria (<104, aerobes/anaerobes and lactobacilli) – BUT with SIBO, the
distal ileum is the transition zone (to dense colonization - >105 BFU per
Diarrhea: Watery (osmotic) – due to blunted villi ml of proximal jejunal aspiration)

Resolution: gluten-free diet (improves within 2 weeks) Diagnosis: mainly clinical (can test it by giving patient antibiotics)
-- Quantitative culture of jejunal aspirate (>105 BFU per ml) – requires
Laboratory Findings: anaerobic sample collection
-- anti-tTG antibodies (IgA) -- radioactively labeled xylose breath test (measure radioactively labeled
-- anti-gliadin antibodies (IgA) CO2 due to bacterial fermentation)
-- anti-endomysial antibodies (IgA) -- hydrogen breath test (after administration of oral lactulose/glucose) –
-- anti-reticulin antibodies (IgA) problem is low sensitivity/specificity
(in case of IgA deficiency, also test for IgG antibodies)

Diagnosis: duodenal biopsy to show: (damage most severe at Symptoms:


duodenum, jejunum and ileum less involved) -- Bloating, abdominal pain, diarrhea, weight loss
-- flattening (blunted) of villi -- Vit B12 deficiency (atrophic glossitis, neurological problems)
-- basal crypt hyperplasia (become deeper) -- Anemia (Fe deficiency, Vit. B12 deficiency)
-- increased intra-epithelial lymphocytes -- Small intestinal slash, polyneuropathy

Genetic testing (to check for HLA DQ2-DQ8) Note: Bacteroides not found in jejunum of healthy people
Inflammatory Bowel Disease: a chronic relapsing/recurrent Epidemiology: Extra-intestinal Manifestations of IBD (usually more in Ulcerative Colitis Crohn’s Disease
bouts of inflammation of the bowel, possibly due to an Peak Age of Onset: b/w 15-30 or 60-80 yrs Crohn’s Disease)
abnormal immune response to enteric flora. Gender: 1:1 (sometimes females more) Inflammation Continuous Segmental/Patchy
In General: pattern
Types: can be divided into: B- Crohn’s Disease: TRANSMURAL inflammation 1- aphthous stomatitis (Recurrent ulcers)
1. Ulcerative Colitis 2- arthritis (hands, knees, back - sacroiliitis) Inflammation Colon Anywhere along GI tract
2. Crohn’s Disease Distribution: can affect any segment in the GI tract 3- erythema nodosum Location (especially terminal ileum)
3. IBD-U (unclassified) Small Bowel Alone (33%), Ileocolic (45%), Colon Alone (20%) 4- pyoderma gangrenosum
à Most commonly affected site is the terminal ileum 5- Episcleritis and uveitis Rectal Always Rare
Diagnosis: of exclusion (many differentials to bloody diarrhea Involvement
and abdominal pain). Presentation: IBD-dependent
- Diarrhea (non-bloody) Skin: Erythema Nodosum Mucosa/Wall Superficial (sandpaper-like) – Deep/Transmural
A- Ulcerative Colitis: SUPERFICIAL inflammation - Abdominal pain à RLQ pain (on, off - esp. after eating) leading Joints: Arthritis (PERIPHERAL) Involvement mucosal + submucosal ulcers
to loss of appetite + weight loss Eyes: Scleritis, Episcleritis
Distribution: always involves the rectum in a continuous - Peri-anal disease (abscess + fistulae formation around rectum) Fistulae/ Rare Very common
fashion, never involves the small intestine. IBD-independent (even if IBD dormant/not flaring up) Stricures (since superficial) (since invasive)
- Acute inflammation (with fever)
1. Proctitis (inflammation of the rectum) Skin: Pyoderma Gangrenosum (especially if stoma bag gets
2. Procto-sigmoiditis (inflammation of the rectum + sigmoid infected after colostomy, treat with anti-TNF) Diarrhea Blood + Mucus Non-bloody
Complications:
colon) Joints: Ankylosing Spondylitis, Sacroiliitis (AXIAL) (since colon affected) (affected area is high up)
1) Strictures (after inflammation, fibrosis results in stricture –
3. Left-sided Colitis (inflammation of rectum + sigmoid colon string sign on TI) Eyes: Uveitis
+ descending colon) -- inflammation of anterior chamber Abdominal LLQ pain (Rectum) RLQ pain (ileum)
2) Obstruction (from edema or fibrosis – blocking bowel,
4. Total (pan) Colitis (anything that goes beyond the splenic -- you get ocular pain, photophobia, redness Pain Mimics appendicitis
dilatation – abdominal distention)
flexure) 3) Abscesses (due to CD’s deep fissures in the mucosa, lead to -- slit lamp exam for diagnosis
-- start on treatment (steroids) Inflammation Crypt Abscesses with neutrophils Lymphoid aggregates with
the collection of pus within GI tract due to inflammation) – can
granulomas
Presentation: Relapsing + Remitting either drain: Other: Primary Sclerosing Cholangitis
- Diarrhea (bloody, mucus) -- into lumen of GI tract (easy) -- inflammation of bile duct
Complications Toxic Megacolon Malabsorption
- Abdominal pain à LLQ pain (Rectum) -- back into fissure, leading to fistula formation -- end up getting strictures and obstruction
Carcinoma Nutritional Deficiency
- Tenesmus 4) Malnutrition -- need liver transplant)
Calcium Oxalate Nephrolithiasis
- Systemic (fatigue, malaise, lethargy, nausea) 5) Fistulas (severe and DEEP inflammation burrows through the à shows a beading pattern, dilation before strictures
Fistulas/Strictures
GI tract layers – tunnel forms) Carcinoma (if colonic)
Complications: -- Perianal (with stool inside) Treatment of IBD
1) Colonic Stricture (inflammation, fibrosis, stricture) -- Abdominal Fistula (with stool and bile) Extra-intestinal Primary sclerosing cholangitis Ankylosing Spondylitis
2) Toxic Megacolon (colon is inflamed + dilated, indication for Goals of Treatment
Findings Positive p-ANCA Sacroiliitis
surgery Differential Diagnosis of IBD: 1. To induce remission
Recurrent mouth ulcers Uveitis
3) Dysplasia or Colonic Carcinoma -- Appendicitis 2. To maintain remission
Migratory Polyarthritis
4) Colonic perforation (severe bleeding in ~1%) -- Diverticulitis 3. To maintain adequate nutrition (or else more inflammation)
Erythema Nodosum
5) Growth Failure (if diagnosed as a kid - short, thin…) -- Carcinoma (if colon affected) 4. To minimize disease- and treatment-related complications
6) Extra-intestinal disorders (arthritis, ocular, renal…) -- Ischemic Colitis 5. To improve the patient’s quality of life
6. With the least quantity of medications possible Effect of Improves Worsens
-- Celiac Disease (Malabsorption, Diarrhea) Smoking (protects against UC) (Increases risk for Crohn’s)
Disease Presentation: symptoms can be divided into:
1) Sx targeted to UC (bowel movement, blood in stool) Etiology and Pathogenesis Ulcerative Colitis
Gross Pseudo-polyps Cobblestone mucosa
2) Sx targeted to systemic inflammation (fever, tachycardia, IBD is caused by an interaction between those three factors: -- Surgery is curative but most of the times we try with medical Appearance Loss of Haustra Creeping Fat
anemia – sign of chronicity, ESR -- inflammatory marker, not 1) Genetic Susceptibility: (most people don’t have family history) therapy before proceeding to surgery
Strictures
used that much anymore – we use CRP these days) -- Race: White >> Black > Asian -- Exceptions: Colon dysplasia/cancer or medically refractive
3) Endoscopy -- Ethnicity: Jew (Ashkenazi) > Jew (Sephardic) > Non-Jew colitis (these are people we directly refer to surgery, risks of
Medications for IBD
-- Twins: Monozygotic (67% CD, 20% UC) > Dizygotic keeping colon)
1) 5-aminosalicylic acid (5-ASA)
Mild Moderate Severe -- Genes: NOD2 explains 20% of CD -- UC (mild-moderate): induce and maintain remission in 50-70% of mild to moderate UC
-- Family History: (positive) increases risk Crohn’s Disease
-- CD: not using them much
Bowel <4 per day 4–6 per day >6 per day -- Neither medical nor surgical therapy provides a cure
2) Glucocorticoids: IV or oral prednisone used in moderate to severe UC and CD. Induce remission.
movements
2) Immune Dysregulation: -- Surgery (to remove inflamed area) is NOT a failure – they
NO role in maintaining remission. Not liked/used due to side effects (bone loss…)
Blood in stool Small Moderate Severe -- Normally: the mucosal immune system controls/down- will feel much better!! 3) Purine Analogues: Azathioprine and mercaptopurine used in glucocorticoid dependent IBD.
regulates gut inflammation in response to luminal contents 4) Methotrexate/Cyclosporine
Fever None <37.5°C mean >37.5°C mean
-- IBD patients: there is an inappropriate and exaggerated Surgical Therapy 5) Biologic agents: (VERY good) anti-TNF agents, anti-IL12/23, anti-integrins
Tachycardia None <90 mean pulse >90 mean pulse mucosal immune response to environmental factors (most Indications for surgery include complications such as:
probably the bacteria) due to failure to down-regulate. • intra-abdominal abscess IBD Surgeries
Anemia Mild <75% of cases > 75% of cases
• medically intractable fistula UC Surgery: Total Colectomy (TAC/IPAA)
Sedimentatio <30 mm >30 mm
3) Environmental Triggers • fibrotic stricture with obstructive symptoms
n rate -- infections • toxic megacolon CD Surgery:
-- anti-biotics (esp. in kids; changes flora, predisposing factor) • intestinal perforation, hemorrhage 1. Ileocecal/Ileocolonic Resection (ICR) then connect via primary anastomosis
Endoscopic Erythema, Marked erythema, Spontaneous
appearance decreased coarse bleeding, -- NSAIDs (horrible because they clamp down blood vessels, lead • dysplasia/cancer 2. Segmental Resection (cut inflamed segment, connect it via anastomosis)
vascular granularity, ulcerations to ulcerations, severe inflammation ) • Patients refractory to/dependent on glucocorticoids 3. Stricturoplasty (to preserve as much of the gut as possible)
pattern, fine contact bleeding,
granularity no ulcerations
-- Diet, SMOKING 4. Diverting Ileostomy (2 stages, 1st – cut colon from cecum, cut sigmoid colon, keep rectum and
-- Stress (very important factor) add ileostomy bag; 2nd – J-pouch 6 months later)
Diverticular Disease
-- Prognosis: Treatment of IBS:
Diverticulosis: (TRUE) outpouching/herniation (1+) of the colonic
- After acute syndrome resolves, patient should have a colonoscopy - Education/Reassurance
mucosa and submucosa through weak spots in the muscular layer of the
(after 8 weeks, to exclude malignancy in case obstruction by tumor) - Dietary Modification:
colonic wall.
- risk of diverticulitis increases after each episode -- avoid short chain CHO b/c osmotically active + poorly absorbed
- Most patients improve over 2-3 days (after antibiotics) -- avoid gas-producing foods
Location: Along any hollow organ (small intestine, stomach…) –
- Surgery recommended if 2-3 episodes of diverticulitis -- avoid gluten and lactose
especially affecting the sigmoid colon (due to higher pressure).
- Physical Activity
à In Asians, right sided more common
-- Treatment: - Probiotics
1. Mild Uncomplicated Attack (1st): - Anti-spasmodics (if patient has abdominal pain)
Pathophysiology: due to high intraluminal pressure weak spots in the
- Outpatient unit, discharged on clear liquid diet and ADAT - Rifaximin (Normix, antibacterial – especially for IBS-D)
muscular layer (especially where vasa recta penetrates the bowel wall)
- use broad-spectrum antibiotics (ciprofloxacin, metronidazole) - Celium Fiber (especially IBS-C, to get water into lumen, and IBS-D to
- colonoscopy done to rule out cancer in that area bulk up their stool)
Epidemiology: affects western societies more, secondary to diet (more
constipation, more pressure, sigmoid has to contract more)
2. Sensitive: if patient cant tolerate PO antibiotics/doesn’t improve:
-- uncommon in patients < 40 years
- Hospitalization w/ CT scan to rule out abscess/perforation Colorectal Cancer Epidemiology (3rd most common cancer in US, 2nd
-- 80% of people > 80 years have diverticulosis
- broad-spectrum antibiotics IV leading cause of neoplastic death)
-- Risk factors:
- percutaneous drainage of abscess + CT/surgery after 6 weeks
1. Increasing age
• Mean Age: 73 years
2. Obesity
3. Peritonitis/Uncontrolled Sepsis/Perforation: Emergency Surgery
3. Constipation
• Pattern:
4. Diet (low fiber, high meat)
4. Fistula repair/stricture surgery/Recurrent diverticulitis: Elective -- Overall incidence decreasing, young people incidence increasing
5. Connective Tissue disorder like Marfan or Ehlers Danlos
-- Men develop CRC 5-10 years earlier than women
syndrome (colon very stretchy)
-- Black people develop CRC 5-10 years earlier than white people
6. Genetic Predisposition
2. Painful Diverticular Disease – abdominal pain with altered bowel
movements with diverticulosis without any other explanation • Risk Factors:
Clinical Findings:
-- IBD (Ulcerative Colitis or Crohn’s Disease)
-- PAINLESS condition without any symptoms or chronic infection
~ related to DURATION of disease and AMOUNT of bowel inflamed
3. Bleeding (most common): PAINLESS, NOT related to diverticulitis ~ related to concomitant primary sclerosing cholangitis (PSC)
Complications:
- patients with diverticulosis have acute PAINLESS lower GI bleeding -- Family History (always screen kid 10 years younger than age of
1. Diverticulitis (more common) – PAINFUL inflammation or infection of
- these bleeds are SELF-LIMITING (but still give blood transfusion) relative affected with CRC, or at least by age 40)
1+ diverticula – could be uncomplicated or complicated (w/abscess,
- perform a COLONOSCOPY to rule out other causes of GI bleeding -- Strep bovis bacteriemia or endocarditis (colonoscopy)
fistula, perforation)
1st – exclude malignancy -- Defined inherited syndromes (small percentage of CRC)
2nd – exclude celiac disease
-- Pathophysiology: due to obstruction of diverticulum by:
3rd – exclude angiodysplasia
~ Fecalith
( diverticulosis itself does not cause anemia unless too much bleeding)
~ Bacterial Flora
~ Micro/Macro-perforation
à if the neck is obstructed, it distends, leads to bacterial overgrowth,
and possible micro-perforation and inflammation. Irritable Bowel Syndrome
It is a functional GI syndrome characterized by chronic abdominal pain
-- Clinical Findings: and change in bowel habits in the absence of any organic cause.
- LLQ abdominal PAIN (which may radiate)
- rebound tenderness (+ over surrounding area) + local guarding Diagnosis: Rome IV criteria = recurrent abdominal pain at least 1
- altered bowel habits (diarrhea usually) day/week in the last 3 months associated with 2+ of the following:
- Anorexia, fever (systemic symptoms) 1. Pain related to defecation
- NO bleeding! 2. Pain associated with a change in stool FREQUENCY
3. Pain associated with a change in stool FORM/APPEARANCE
-- Complications: Should exclude presence of a more serious illness (anemia, occult…)
1. Abscess formation (collection of pus + neutrophilic inflammation)
2. Rupture of diverticulum (leads to perforation + peritonitis – Epidemiology: females are diagnosed more than 2x as often as males
especially in the elderly/immunosuppressed)
3. Colonic Obstruction (in case of repeated episodes) Subtypes:
4. Pyelophlebitis (in patients with jaundice + hepatic abscesses) 1- IBS with predominant constipation (IBS-C)
5. Fistulas (when abscess opens next to an organ) 2- IBS with diarrhea (IBS-D) – associated with increased serotonin
3- Mixed IBS
-- Diagnostic Studies: (no endoscopy – can perforate) 4- Unclassified IBS
1. CBC shows leukocytosis
2. Urine analysis shows pyuria Clinical Features: MUCUS with stools, PERIODIC + abdominal pain
3. Ultrasonography shows inflammation (not really good, operator (not bloody, no large volume, they do NOT wake up at night, no weight
dependent, guarding and rebound pain make it hard) loss, no greasy stools, no anemia…)
4. CT scan can be useful to detect abscess (20% false –ve)
5. Enema studies identify diverticulosis but not diverticulitis
I- Functions of the liver: Cannot survive without liver; it is involved in: Hepatocyte Structure: hepatocytes have three membranes in three Hepatic Stellate (Ito) Cell Function: (occupy space of Disse) Hepatic Microcirculation: (it’s a LOW PRESSURE system)
1- Metabolism, detoxification, and inactivation of endogenous (bilirubin, different planes (not touching) They are cells that store fat and vitamin A. Flow at low pressure is essential for the exchange that takes place
ammonia) AND exogenous (drugs) substances 1- BASOLATERAL Sinusoidal Memrbane (contains microvilli for They play an active role in: continuously between hepatocytes and blood.
2- Kupffer cells of liver involved in filtering blood of foreign substances (virus exchange with plasma, in direct contact with sinusoids) • Regulation of microvascular tone and blood flow, hence portal
infected cells, tumor cells…) 2- APICAL Cannalicular Membrane (where cell secretes bile into the pressure The normal portal pressure is < 7 mmHg.
3- Conversions of important hormones and vitamins into active forms, e.g., biliary canaliculus) • Regeneration of hepatocytes
hydroxylation of vitamin D, and de-iodination of T4 into T3 3- LATERAL Desmosomal Membrane (faces other hepatocytes) • Fibrogenesis (transformation into myofibroblasts) when Portal Hypertension: (pressure goes > 10 mmHg)
4- Procession of lipophilic chemicals into water soluble forms (cannot be activated by inflammation, alcohol…
processed in kidney) Hepatocyte Function: (organelles) Portal hypertension may have a number of different causes:
5- Synthesis of bile salts and bile secretion Activation of Ito cells is at the center of liver fibrosis -- Anytime 1) Pre-hepatic:
6- Storage of proteins, carbohydrates, lipids and minerals Mitochondria – energy generation hepatocytes die (apoptosis or necrosis), Ito cells and Kuppfer cells • Schistosomiasis
7- Synthesis of essential proteins (like albumin), carbohydrates, metabolites • oxidative phosphorylation are activated leading to TGF-beta release and fibrosis (deposition of • Portal Vein Thrombosis
8- Release of nutrients to other organs, in the free (e.g., glucose – important • b-oxidation of fatty acid collagen in liver sinusoids) => CENTRAL mechanism to keep in • Malignant Tumors
for brain) or bound form (e.g., triglyceride) • heme synthesis mind
• enzymes for citric acid cycle 2) Intra-hepatic
Strategic location of the liver as the only organ b/w splanchnic and Liver Regeneration: (begins in zone I, oxygenated, richer) • Cirrhosis
systemic circulation enables functions RER The liver is capable of regeneration; its an organ of conditional à Alcohol
• Protein synthesis (albumin – maintains oncotic pressure, renewal: à Chronic Hepatitis
II- Functional Anatomy: the liver is functionally divided into segments, each carries endogenous and exogenous molecules like lipids, -- at baseline, hepatocytes and cholangeocytes die but are replaced • Congenital Hepatic Fibrosis
with their own independent blood supply (hepatic artery, portal vein) and coagulation factors) slowly
independent biliary drainage. • Triglyceride synthesis, complexing to lipoproteins -- during ongoing liver injury (rapid tissue loss) it’s a very high level 3) Post-hepatic
• Glucose-6-phosphatase synthesis – vital to generate glucose replacement (from Stem cells: oval cells + bone marrow cells) • Hepatic Vein Thrombosis (Budd-Chiary Syndrome)
How is this division made? like when fasting • Right-sided heart failure
1- Draw a line b/w right and left lobes (connect IVC and gallbladder) so the Liver Injury: They require adequate amount of growth factors and • Constrictive Pericarditis
left lobe would consist of the caudate + quadrate lobes. SER primers high/fast replacement UPON need. In case destruction rate
2- Divide each lobe into medial and lateral sectors • Bilirubin conjugation overpowers them, they begin proliferating in a disorderly fashion Consequences of portal HTN include:
3- Divide each sector into anterior and posterior subsector • P-450 systems (detoxification) (associated with fibrosis, and eventually liver cirrhosis). 1) Collaterals between the portal and the systemic circulation (leading to
Thus, a functional unit may be surgically removed (segmentectomy, for • Steroid, cholesterol, bile acid synthesis increased blood flow in these veins):
example, if it has a cancer) without affecting the function of other units. • Induced by phenobarbital Genetic Regulators: Proliferation involves c-fos, c-jun, c-myc, NO • Azygos à Gastro-esophageal varices (+/I hemorrhage)
synthase, HGF, EGF, TGF-α and β • Umbilical à Caput medusae (reopening of umbilical vein) –
III- Functional Unit of Liver: Rappaport’s (portal) Acinus Lysosomes converges on the umbilicus, not like IVC thrombosis
It consists of a 3D mass of hepatocytes with two axes: • Hydrolytic enzymes for cell destruction (filtering) Minimum Amount: What is the minimum amount of liver you need • Retroperitoneal system à retroperitoneal varices
- Axis going through two portal triads • Deposition of ferritin and copper • Rectal system à huge hemorrhoids and rectal varices
for regeneration (sufficient critical mass of hepatocytes for
- Axis going through two central veins regeneration)? 2) Formation of ascites
Golgi: Packaging and excretion of proteins/waste products into bile • Minimum amount is 20% (normal) 3) Splenomegaly
Hepatocytes from different zones are genetically, structurally, and functionally or into circulation • Minimum amount is 30% (mild/controlled disease) 4) Hepatic encephalopathy (liver not functional, ammonia cannot be
different. • Minimum amount is 40% (cirrhotic liver) processed to urea, travels to brain)
Zone I Hepatocytes = next to periportal area (hepatic artery, portal vein) Endothelial Cell Structure: the endothelial cells lining the sinosoids
-- better oxygenated (less susceptible to hypoxia) are organized in a discontinuous manner (not continuous/tight, like Hepatic Blood Flow: The most serious associated condition is liver cirrhosis.
-- larger + more numerous mitochondria (more Kreb’s cycle enzymes) in capillaries) with multiple fenestrations (pores) in between
-- more active in bile secretion allowing for the exchange of material between hepatocytes and Liver receives about 1.5 L/min of blood flow (almost a ¼ of CO, and Pressure of Vascular Bed: determined by two main factors:
-- more fenestrations of surrounding endothelial cells plasma (no basement membrane). blood content constitutes a third of the liver’s total weight) 1) Resistance
-- less numerous smooth ER, less P450 (more susceptible to toxins) -- 2/3 is from portal vein 2) Flow Rate
-- more susceptible to viral hepatitis (due to close proximity to portal blood) Pressure in Sinosoids: 2-3 mmHg -- 1/3 is from hepatic artery
(Gluconeogenesis, Cholesterol Synthesis, Ureagenesis, Beta Oxidation) Pressure in Capillaries: 18-19 mmHg (bile duct receives blood from branches of right hepatic artery)

Zone III Hepatocytes = closest to hepatic vein Endothelial Cell Function: Hepatic Arterial Buffer Response:
-- less oxygenated (more susceptible to hypoxia) -- Lipid, Vitamin A, and cholesterol homeostasis If portal vein blood flow is reduced (portal vein thrombosis due to
-- smaller + less numerous mitochondria -- Filtration of blood (wastes, clear pathogens and harmful prolonged OCP use), hepatic artery is dilated to compensate. This
-- less fenestrations of surrounding endothelial cells enzymes) process is mediated by adenosine.
-- more numerous smooth ER, more P450 (less susceptible to toxins)
-- fat accumulation begins here What’s the difference between liver sinusoids and systemic Liver Congestion: In conditions where systemic venous pressure is
-- processes taking place: capillaries? high (congestive right sided heart failure), both hepatic venous and
(Glycogen Synthesis, Glycolysis, Lipogenesis, Ketogenesis, Glutamine The liver sinusoids have: portal venous radicles dilate, a process that leads to “liver
Formation (to remove ammonia), Biotransformation of drugs (detox)) 1. No tight junctions congestion”, and ultimately might lead to cardiac cirrhosis (cardiac
2. No basement membranes hepatopathy).
3. Fenestrations (to allow exchange with hepatocytes)
Blood Flow
Ischemic Hepatitis: A sudden drop in blood pressure, leads to
Portal Hypertension: In liver disease, Ito cells are activated and constriction of both hepatic venous and portal venous radicles. This
Hepatic Bile Portal Zone I Zone II Zone I Hepatic turn into fibroblasts that deposit collagen in the sinosoids. The leads to marked diversion of blood flow away from the liver, with
à à (Periportal) (Midzone)
Artery Duct Vein (Centrilobular) Vein vessels are thus obliterated and capillarated. They become an area resultant liver ischemia, necrosis.
of high pressure (since resistance increased, and pressure = flow x
resistance). This forms the root of portal hypertension.
Bile Secretion

You might also like