Skip to content

Abdominal Pain

Abdominal pain is a prevalent and often challenging symptom characterized by discomfort or distress originating from the abdominal region. It can manifest as acute or chronic, stemming from a diverse range of underlying conditions affecting various organ systems within the abdomen. Understanding its varied presentations and etiologies is fundamental for accurate diagnosis and effective management.

The biological basis of abdominal pain involves complex physiological pathways and genetic predispositions. Research indicates that genetic factors contribute to an individual’s susceptibility to chronic pain, with SNP-based heritability estimated at 6.9% on the observed scale and 12.3% on the liability scale[1]. Genome-wide association studies (GWAS) have been instrumental in investigating associations between genetic variations and the susceptibility to chronic pain, including analyses of specific subgroups. These studies aim to uncover the intricate mechanisms underlying pain perception and chronicity.

Clinically, abdominal pain is a frequent reason for individuals to seek medical attention, presenting a significant diagnostic challenge due to its wide spectrum of potential causes. The distinction between transient discomfort and persistent, chronic pain is critical for patient care, as chronic conditions can profoundly impact quality of life. The ongoing advancement in understanding the genetic underpinnings of pain holds promise for developing more precise diagnostic tools and personalized treatment approaches.

The social importance of addressing abdominal pain is considerable, impacting public health, healthcare systems, and individual productivity. Chronic abdominal pain can lead to substantial disability, economic burden, and reduced overall well-being. Large-scale genetic meta-analyses, involving hundreds of thousands of individuals, are conducted to achieve higher statistical power for studying pleiotropic effects and genetic correlations of pain, ultimately aiming to improve prevention and treatment strategies[1].

Study Design and Statistical PowerResearch on abdominal pain, particularly in genetic association studies, faces limitations related to statistical power and study design. Many studies are underpowered to achieve genome-wide significance (GWS), often due to relatively small sample sizes in discovery and replication cohorts. This can lead to findings that do not reach GWS, and in some cases, observed effect sizes may be larger than those reported in other pain GWAS, potentially indicating inflated associations. For instance, statistical power for specific genetic models may be moderate, even for relatively large observed effect sizes, underscoring the challenge of robust discovery in complex traits.

Furthermore, current genotyping platforms do not capture all known common genetic variations across the human genome, representing only about two-thirds of them [2]. This incomplete genomic coverage can increase the risk of false discoveries and limit the comprehensive identification of genetic factors. Replication of findings with larger sample sizes by independent investigators is crucial to confirm novel associations, especially given instances where variants initially identified in discovery cohorts do not replicate in additional groups, leading to weak evidence for association.

Phenotypic Assessment and Population SpecificityA significant limitation in understanding abdominal pain genetics stems from the challenges in phenotypic measurement. Studies often rely solely on participants’ self-assessment of pain severity, which, while common in clinical practice, can introduce unreliability. This unreliability arises from the use of non-standardized or unvalidated survey instruments and the potential for recall bias. Such methodological issues can lead to reporting errors and may bias effect size estimates, impacting the accurate interpretation of identified genetic associations.

Moreover, the generalizability of findings is frequently restricted, as many studies focus exclusively on specific populations, such as European Americans [3]. Pain responses, including analgesic efficacy and underlying genetic variations, are known to differ significantly among various ethnic populations[3]. Therefore, results from one ancestral group cannot be broadly applied to others, highlighting the critical need for research that includes diverse ethnic backgrounds to ensure broader applicability and understanding of abdominal pain genetics.

Incomplete Genetic Architecture and Mechanistic UnderstandingDespite advancements, the genetic architecture of abdominal pain remains incompletely understood, reflecting a broader challenge in complex trait genetics. Even for traits with estimated heritability, significant genetic variants identified through genome-wide association studies (GWAS) often explain only a small fraction of individual differences, pointing to substantial “missing heritability.” This suggests that many genetic factors, or their interactions, are yet to be discovered.

Genetic association studies are primarily designed to identify statistical relationships, not to elucidate the underlying biological mechanisms. Therefore, follow-up studies are essential to characterize how identified genetic loci contribute to abdominal pain, particularly when candidate genes lack clear functional annotation. Extensive additional research in both animal models and human subjects is required to translate statistical associations into a comprehensive understanding of the biological pathways involved in pain perception and processing.

The GRB10gene, or Growth Factor Receptor Bound Protein 10, encodes an adaptor protein crucial for regulating cellular growth and metabolism. It primarily functions by interacting with and modulating the activity of various receptor tyrosine kinases, particularly the insulin receptor and insulin-like growth factor 1 (IGF-1) receptor. Through these interactions, GRB10 influences key pathways involved in cell proliferation, differentiation, and nutrient sensing, playing a role in processes from embryonic development to adult metabolic homeostasis. Its expression is complex, often exhibiting genomic imprinting, meaning its activity can be influenced by whether the gene copy was inherited from the mother or father.

The single nucleotide polymorphism (SNP)rs4947737 is located within the GRB10 gene, potentially impacting its regulatory or coding regions. Variants like rs4947737 can alter gene expression levels, affect the stability of the messenger RNA, or even modify the structure or function of the GRB10 protein itself. Such changes could lead to subtle yet significant shifts in the efficiency of insulin or IGF-1 signaling pathways. Consequently, these genetic variations may influence how cells respond to growth factors and metabolic cues, affecting a wide range of physiological processes.

Variations in GRB10, such as rs4947737 , hold potential implications for conditions involving abdominal pain due to the gene’s central role in metabolic regulation and cell signaling. Dysregulation of insulin and IGF-1 pathways can contribute to inflammation, altered gut motility, and visceral hypersensitivity, all of which are underlying mechanisms of various abdominal pain syndromes like irritable bowel syndrome (IBS) or inflammatory bowel diseases. By influencing these fundamental biological processes,rs4947737 could modulate an individual’s susceptibility to or experience of abdominal pain, potentially by affecting neural signaling in the gut or the body’s overall inflammatory response.

RS IDGeneRelated Traits
rs4947737 GRB10abdominal pain

Classification, Definition, and Terminology

Section titled “Classification, Definition, and Terminology”

Definition:Abdominal pain is defined as discomfort or aches reported in the stomach or abdomen area. It is identified as one of several distinct body sites where pain can be experienced[4].

Classification:Abdominal pain can be classified based on its duration. When pain reported in the stomach or abdomen has persisted for three months or longer, it is categorized as chronic[4]. This site-specific pain (e.g., in the stomach/abdomen) is distinct from pain reported as “all over the body”[4].

Terminology:

  • Stomach/Abdomen Pain:This specific term refers to pain localized to the abdominal region, as identified in participant questionnaires[4].
  • Chronic Pain:This classification applies to any pain, including abdominal pain, that has a duration of three months or longer[4].
  • Body Mass Index (BMI):A measure of body fat based on an individual’s height and weight. Elevated BMI is a recognized risk factor for chronic widespread pain (CWP), a condition that frequently co-occurs with functional somatic syndromes such as irritable bowel syndrome (IBS), which often presents with abdominal pain[4].
  • Gut Microbiome:The collection of microorganisms inhabiting the human gut. Its composition is influenced by various factors including diet, age, sex, and body weight, and it plays a significant role in human health and disease. Research indicates that CWP, which can co-occur with IBS, is associated with decreased alpha diversity of the gut microbiome[4].
  • Healthy Eating Index (HEI):A scoring system, ranging from 0 to 100, developed by the United States Department of Agriculture (USDA) to evaluate the quality of a diet based on its alignment with dietary recommendations. Diet is a known factor influencing the gut microbiome, which is relevant to conditions like CWP and related functional somatic syndromes[4].

Abdominal pain is typically described as discomfort or pain localized to the stomach or abdomen[5]. It can manifest in various ways, with different qualities, durations, and severity levels.

Abdominal pain can be identified as a specific body site of pain[5]. When it persists for three months or longer, it is classified as chronic pain[5]. The nature of the pain can be assessed through qualities such as burning, tingling, sudden onset, and sensitivity to heat and cold[5].

In the context of gastrointestinal autoimmune disorders, such as inflammatory bowel disease, abdominal pain may present as “on-going acute” rather than strictly chronic. This type of pain can result from physical issues like stricture, abscesses, or partial or complete obstruction of the small bowel[6].

Abdominal pain is commonly assessed through self-reported questionnaires. Participants may select “stomach/abdomen” as a site of pain on a touchscreen questionnaire[5]. The duration of pain is also a key factor, with individuals asked if their pain has lasted for three months or longer to determine chronicity[5].

Severity is often measured using ordinal scales. For instance, dysmenorrhea pain severity, a type of abdominal pain, has been captured using a four-point scale with options ranging from “not painful” to “extremely painful”[7].

To characterize the nature of pain, questionnaires like the painDETECT questionnaire are used. This seven-item tool employs a Likert scale to gather descriptions of pain qualities, including burning pain, tingling, sudden pain, and sensitivity to heat and cold, which helps distinguish different pain types[5].

Abdominal pain can vary significantly among individuals. It is often distinguished from widespread “pain all over the body” by its specific localization to the stomach or abdomen[5]. The duration of pain is also variable, with a clear distinction made between acute and chronic presentations, the latter defined by a duration of three months or more[5].

The severity of abdominal pain can also differ widely, as evidenced by the distribution of responses on severity scales for conditions like dysmenorrhea[7]. Furthermore, the underlying mechanisms and presentation of pain can vary; for example, pain associated with gastrointestinal autoimmune disorders may be driven by distinct genetic variations and mechanisms involving the enteric nervous system, leading to an “on-going acute” rather than a purely chronic experience[6].

Abdominal pain can stem from a variety of genetic and environmental factors. In some cases, such as with gastrointestinal autoimmune disorders like ulcerative colitis (UC), inflammatory bowel disease (IBD), and Crohn’s disease, the pain might be characterized as “on-going acute” rather than chronic. This type of pain often results from structural issues within the small bowel, including strictures, abscesses, or partial or complete obstruction[8].

Genetic influences play a role in the predisposition to and experience of pain, which can include abdominal pain. Chronic pain, in general, has been observed to be heritable within extended families[9]. Research suggests that a single underlying genetic factor may explain pain reported across different body sites[10]. Specific genetic risks for chronic pain have been identified, including contributions from risk variants associated with major depressive disorder[11]. Studies on chronic low back pain, a form of pain that can be related to the abdominal region, indicate a role for heritability[12]. Furthermore, genome-wide meta-analyses have pinpointed specific genetic loci linked to chronic back pain[1].

Environmental elements also contribute to the development and experience of abdominal pain. For chronic pain broadly, environmental risks are recognized[11]. Lifestyle factors, for instance, are associated with chronic low back pain[12]. In the specific context of menstrual pain, which is a type of abdominal pain, disruptions in social relationships can intensify the link between emotional distress and pain in young women[13].

Abdominal pain arises from a complex interplay of biological mechanisms, involving structural, molecular, and cellular pathways.

In gastrointestinal autoimmune disorders such as Ulcerative Colitis, Inflammatory Bowel Disease, and Crohn’s Disease, abdominal pain can be linked to physical alterations within the digestive system. These alterations include the development of strictures, abscesses, and partial or complete obstruction of the small bowel[8]. The pain experienced in these conditions may be described as ongoing acute rather than chronic[8].

Inflammatory MediatorsProstaglandins are important inflammatory molecules that contribute to pain signaling. For example, specific inhibitors of cyclooxygenase-2 (COX-2) are used in the management of dysmenorrhea, suggesting the involvement of COX-2-driven prostaglandin production in menstrual pain[14].

Extracellular Matrix and Neuropathic PainStudies indicate that the organization of the extracellular matrix is a significant factor in both inflammatory and neuropathic pain[15]. Research also focuses on identifying potential mechanisms and central genes involved in neuropathic pain[16].

Abdominal pain arises from a complex interplay of molecular and physiological mechanisms involving the nervous system, immune system, gut microbiome, and genetic factors.

The gut-brain axis (GBA) facilitates communication between the central nervous system (CNS) and the enteric nervous system (ENS), which can operate somewhat independently from the CNS [17]. This axis plays a significant role in pain perception. The nociceptive signaling system, responsible for processing pain, can undergo alterations, potentially disrupted by excessive stress responses[6]. Structural and functional changes in the brain are associated with the transition to chronic pain, although for gastrointestinal autoimmune disorder-associated pain, the ENS and GBA may play a more central role than CNS changes[6]. Heightened brain responses to pain anticipation have been observed[16].

Inflammation is a key driver of abdominal pain, particularly in conditions like inflammatory bowel diseases (IBD). In IBD, pain can stem from structural issues such as strictures, abscesses, and partial or complete obstruction of the small bowel[6]. The immune and nervous systems interact directly in the experience of pain[18]. Molecular mediators like cytokines and chemokinesare crucial at the intersection of neuroinflammation, neurodegeneration, and neuropathic pain[19]. Macrophages, immune cells involved in inflammation, contribute to metabolic diseases and insulin resistance[20]. The adipose tissue’s expression of tumor necrosis factor-alpha (TNF-alpha), a pro-inflammatory cytokine, also plays a direct role[21].

The human gut microbiome significantly influences human health and disease[22]. Its composition is affected by host genetics, diet, age, sex, weight, medication use, and other environmental factors[22]. In turn, the gut microbiome can impact host metabolism[23]. While a portion of the gut microbiome is heritable[24], environmental factors are understood to have a greater effect on its overall shaping [25]. The microbiome exhibits temporal variability, which is a personalized feature [26]. Research indicates a connection between chronic widespread pain and the gut microbiome[27].

Genetic determinants contribute to the development of chronic pain, with some shared genetic factors identified between chronic widespread pain, low back pain, and frailty[6]. Genetic variation and mechanisms underlying chronic pain associated with gastrointestinal autoimmune disorders (such as ulcerative colitis, IBD, and Crohn’s Disease) may differ from those associated with pain outside the digestive system[6]. Epigenetic modifications, specifically DNA methylation in neurological pathways, are implicated in chronic widespread pain[6]. Host genetics also play a role in shaping the gut microbiome[24].

The body’s internal biological clock, or circadian rhythm, exerts control over pain and neuroinflammation[28]. The diurnal and twenty-four hour patterning of human diseases, including acute and chronic conditions, highlights the influence of these rhythms [29].

Metabolic factors, such as overweight, obesity, and a raised Body Mass Index (BMI), are established risk factors for chronic widespread pain[6]. Longitudinal studies suggest that an elevated BMI often precedes the onset of pain, rather than being a consequence of pain-induced immobility[6]. Increased fat mass is also associated with these conditions [6].

Abdominal pain is a common symptom with diverse underlying causes, and understanding its specific origins has significant clinical implications for diagnosis, prognosis, and management.

In the context of gastrointestinal autoimmune disorders like ulcerative colitis (UC), inflammatory bowel disease (IBD), and Crohn’s disease, abdominal pain may be characterized as “on-going acute” rather than chronic. This distinction is crucial because the pain often results directly from structural complications such as strictures, abscesses, or partial/complete obstruction of the small bowel[6]. The genetic variations and mechanisms underlying this type of pain appear distinct from those associated with chronic pain conditions outside the digestive system[6]. Furthermore, the role of the enteric nervous system (ENS) operating independently from the central nervous system (CNS), along with the gut-brain axis (GBA), suggests that structural and functional brain changes typical of chronic pain may be less central in these gastrointestinal conditions[6]. This implies that therapeutic approaches might need to target the peripheral mechanisms and gut-specific pathways more directly.

Another critical condition associated with abdominal discomfort is Abdominal Aortic Aneurysm (AAA). Early diagnosis of AAA is vital, as studies indicate that a significant proportion of individuals with AAA are diagnosed late in their disease course[30]. This highlights the underutilization of screening methods for AAA, despite recommendations from bodies like the US Preventive Services Task Force [31]. Population-level screening has been shown to influence the incidence of ruptured AAA, underscoring its prognostic value [32]. Research also demonstrates that population risk factor estimates for AAA can be effectively derived from electronic medical records, which could aid in identifying at-risk individuals [33]. The development of genome-wide polygenic risk scores (PRS) for AAA also holds promise for identifying individuals at higher genetic risk, potentially guiding screening efforts and preventive strategies.

Frequently Asked Questions About Abdominal Pain

Section titled “Frequently Asked Questions About Abdominal Pain”

These questions address the most important and specific aspects of abdominal pain based on current genetic research.


1. My family all gets stomach aches; will I definitely get them too?

Section titled “1. My family all gets stomach aches; will I definitely get them too?”

While there’s a genetic component to chronic pain, including abdominal pain, it doesn’t mean you’ll definitely experience it. Studies suggest genetic factors contribute about 7-12% to your susceptibility to chronic pain, indicating a predisposition, but many other factors are at play.

2. Why do some people just seem to handle stomach pain better than me?

Section titled “2. Why do some people just seem to handle stomach pain better than me?”

Your individual genetic makeup can influence your pain perception and tolerance. Variations in genes contribute to these differences, meaning some people are biologically predisposed to experience pain more intensely or frequently than others.

Yes, research indicates that pain responses and underlying genetic variations can differ among various ethnic populations. Many studies have focused on specific groups, so your ancestral background might influence your specific genetic risk factors for abdominal pain.

4. Can eating healthy help reduce my stomach pain if it’s genetic?

Section titled “4. Can eating healthy help reduce my stomach pain if it’s genetic?”

Yes, even with a genetic predisposition, lifestyle choices like healthy eating can be beneficial. Genes likeGRB10influence metabolic pathways, such as insulin signaling, which are impacted by diet, potentially modulating your body’s overall response to pain.

5. Does my metabolism have anything to do with my constant stomach issues?

Section titled “5. Does my metabolism have anything to do with my constant stomach issues?”

There can be a connection. Genes such as GRB10play a role in regulating cellular metabolism and how your body responds to growth factors like insulin. Variations in these genes can subtly affect these metabolic processes, which might influence your susceptibility to abdominal pain.

6. Is getting a DNA test useful to figure out my stomach pain?

Section titled “6. Is getting a DNA test useful to figure out my stomach pain?”

A DNA test might identify certain genetic variations linked to pain susceptibility, like those in theGRB10gene. However, genetics currently explain only a fraction of abdominal pain risk, and many factors remain undiscovered, so it’s not a full diagnostic tool yet.

7. Is my stomach pain all in my head, or is there a real cause?

Section titled “7. Is my stomach pain all in my head, or is there a real cause?”

Abdominal pain is very real and has a clear biological basis, involving complex physiological pathways and genetic predispositions. Genetic factors contribute significantly to your susceptibility to chronic pain, meaning there’s a tangible biological foundation, not just a psychological one.

8. Why does my stomach pain keep returning even after treatment?

Section titled “8. Why does my stomach pain keep returning even after treatment?”

Chronic pain often has a genetic component, which can predispose you to persistent symptoms. Your genetic makeup influences how your body processes pain signals, making it more likely for discomfort to recur even after initial interventions, due to an underlying biological susceptibility.

9. Why is it so hard for doctors to find my stomach pain cause?

Section titled “9. Why is it so hard for doctors to find my stomach pain cause?”

Abdominal pain is complex with many potential causes, and its genetic architecture is still not fully understood. Even with advanced studies, identified genetic variants often explain only a small part of individual differences, leaving many underlying factors unknown and diagnosis challenging.

10. Can new treatments be made just for mytype of stomach pain?

Section titled “10. Can new treatments be made just for mytype of stomach pain?”

The ongoing research into the genetic basis of pain is very promising for personalized medicine. By understanding specific genetic variations, future treatments could be specifically tailored to your unique biological profile, leading to more effective and targeted therapies.


This FAQ was automatically generated based on current genetic research and may be updated as new information becomes available.

Disclaimer: This information is for educational purposes only and should not be used as a substitute for professional medical advice. Always consult with a healthcare provider for personalized medical guidance.

[1] Suri, P., et al. “Genome-wide meta-analysis of 158,000 individuals of European ancestry identifies three loci associated with chronic back pain.” 2018, vol. 48, no. 7, pp. 1–23.

[2] Pe’er, Itsik, et al. “Evaluating and improving power in whole-genome association studies using fixed marker sets.” Nature Genetics, vol. 38, no. 6, 2006, pp. 663–667.

[3] Edwards, C., Fillingim, R., & Keefe, F. “Race, ethnicity and pain.”Pain, vol. 94, no. 2, 2001, pp. 133–137.

[4] Johnston, Keira J. A., et al. “The gut microbiome is associated with chronic widespread pain.”Communications Biology, vol. 2, no. 1, 2019, p. 321.

[5] Lim, V. K. T., et al. “Genetic architecture of chronic pain in the UK Biobank.”Communications Biology, vol. 3, no. 1, 2020, p. 329.

[6] Smith, M. A. W. T. H., et al. “The genetics of chronic pain: A systematic review and meta-analysis of genome-wide association studies.”PLoS Genetics, vol. 17, no. 10, 2021, e1009804.

[7] Harris, Sarah E., et al. “Genome-wide association meta-analysis of dysmenorrhea reveals two novel loci and a functional link to endometriosis.”Pain, vol. 162, no. 5, 2021, pp. 1528-1537.

[8] Docherty MJ et al. “Managing Pain in Inflammatory Bowel Disease.”Gastroenterol Hepatol (N Y), vol. 7, no. 9, 2011, pp. 592-601.

[9] Hocking LJ, Morris AD, Dominiczak AF, Porteous DJ, Smith BH. Heritability of chronic pain in 2195 extended families. Eur J Pain (United Kingdom). 2012; 16(7):1053–63.

[10] Williams FMK, Spector TD, MacGregor AJ. Pain reporting at different body sites is explained by a sin-gle underlying genetic factor. Rheumatology. 2010; 49(9):1753–5.

[11] McIntosh AM, Hall LS, Zeng Y, Adams MJ, Gibson J, Wigmore E, et al. Genetic and Environmental Risk for Chronic Pain and the Contribution of Risk Variants for Major Depressive Disorder: A Family-Based Mixed-Model Analysis. PLoS Med. 2016; 13(8):1–17.

[12] Junqueira DRG, Ferreira ML, Refshauge K, Maher CG, Hopper JL, Hancock M, et al. Heritability and lifestyle factors in chronic low back pain: Results of the Australian Twin Low Back Pain Study (The AUTBACK study). Eur J Pain (United Kingdom). 2014; 18(10):1410–8.

[13] Alonso C, Coe CL. Disruptions of social relationships accentuate the association between emotional distress and menstrual pain in young women. Health Psychol 2001;20:411–16.

[14] Harel, Z. “Cyclooxygenase-2 specific inhibitors in the treatment of dysmenorrhea.” Journal of Pediatric and Adolescent Gynecology, vol. 17, 2004, pp. 75–79.

[15] Parisien, M., Samoshkin, A., Tansley, S. N., Piltonen, M. H., Martin, L. J., El-Hachem, N., et al. “Genetic pathway analysis reveals a major role for extracellular matrix organization in inflammatory and neuropathic pain.”Pain, vol. 160, 2019, pp. 932–944.

[16] Gu, Y., Qiu, Z., Cheng, N., Chen, C., Hei, Z., & Li, X. “Identification of potential mechanism and hub genes for neuropathic pain by expression-based genome-wide association study.”Journal of Cellular Biochemistry, vol. 120, 2019, pp. 4912–4923.

[17] Carabotti M et al. “The gut-brain axis: interactions between enteric.”

[18] Ren K et al. “Interactions between the immune and nervous systems in pain.”Nat Med, 2010, vol. 16.

[19] Ramesh G et al. “Cytokines and chemokines at the crossroads of neuroinflammation, neurodegeneration, and neuropathic pain.”Mediators Inflamm, 2013.

[20] Chawla A et al. “Macrophage-mediated inflammation in metabolic disease.”Nat Rev.

[21] Hotamisligil GS et al. “Adipose expression of tumor necrosis factor-alpha: direct role in obe-.”

[22] Kurilshikov A et al. “Host genetics and gut microbiome: challenges and perspectives.”Trends Immunol, vol. 38, 2017, pp. 633-47.

[23] Janssen AW et al. “Potential mediators linking gut bacteria to metabolic health: a critical view.”J Physiol, vol. 595, 2017, pp. 477-87.

[24] Goodrich JK et al. “Human genetics shape the gut microbiome.”Cell, vol. 159, 2014.

[25] Rothschild D et al. “Environment dominates over host genetics in shaping human gut microbiota.”Nature, vol. 555, 2018, pp. 210-5.

[26] Flores GE et al. “Temporal variability is a personalized feature of the human microbiome.” Genome Biol, 2014, p. 531.

[27] “Chronic widespread pain and the gut microbiome.”academic.oup.

[28] Segal JP et al. “Circadian control of pain and neuroinflammation.”

[29] Smolensky MH et al. “Diurnal and twenty-four hour patterning of human diseases: Acute and chronic common and uncommon medi-.”

[30] Mell, M. W., Hlatky, M. A., Shreibati, J. B., Dalman, R. L., Baker, L. C. “Late diagnosis of abdominal aortic aneurysms substantiates underutilization of abdominal aortic aneurysm screening for Medicare beneficiaries.”J Vasc Surg.

[31] Owens, D. K., Davidson, K. W., Krist, A. H., Barry, M. J., Cabana, M., Caughey, A. B., Doubeni, C. A., Epling, J. W. Jr, Kubik, M., Landefeld, C. S., et al. “Screening for abdominal aortic aneurysm: US Preventive Services Task Force rec-.”jama.

[32] Scott, R. A., Wilson, N. M., Ashton, H. A., Kay, D. N. “Influence of screening on the incidence of ruptured abdominal aortic aneurysm: 5-year results.”bjs.

[33] Smelser, D. T., Tromp, G., Elmore, J. R., Kuivaniemi, H., Franklin, D. P., Kirchner, H. L., Carey, D. J. “Population risk factor estimates for abdominal aortic aneurysm from electronic medical records: a case control study.”