Lymphocytes: Diagnostic Significance and Clinical Insights
Authors: Payal Bhandari M.D., Emilia Feria, Madison Granados
Contributors: Vivi Chador, Hailey Chin
|
Key Insights
Blood contains three types of cells: red blood cells (RBCs), white blood cells (WBCs), and platelets.
RBCs carry oxygen to tissues using hemoglobin, which gives blood its red color.
WBCs defend the body against infections and foreign invaders like tumor cells, regulate inflammation, and support immune balance.
Platelets help stop bleeding by forming clots, repairing injuries, and aiding in new blood vessel growth.
Figure 1: Blood is made up of 55% plasma (mostly water, with salts, proteins, lipids, and glucose), 45% red blood cells (RBCs), and 1% white blood cells (WBCs) and platelets. A thicker buffy coat, the white blood cell layer, can indicate leukemia, meaning “whiteness of blood” in Greek. Under a microscope, white cells are colorless but are stained with hematoxylin (blue) and eosin (pink) to differentiate them. For example, eosinophils take up the pink stain strongly.
A WBC differential measures the five main types of white blood cells: neutrophils, lymphocytes, monocytes, eosinophils, and basophils. Lymphocytes play a key role in eliminating pathogens, cancer cells, and foreign invaders. Their levels in the blood can reveal health issues like infections, autoimmune disorders, blood cancers, and inflammation. Lifestyle factors such as diet, medications, exercise, hydration, and stress can impact lymphocyte levels, influencing the management of conditions linked to a shorter lifespan.
What Are Lymphocytes?
Lymphocytes, including T cells, B cells, and natural killer (NK) cells, are vital for the immune system. They identify and destroy pathogens like viruses, bacteria, and cancerous or damaged cells 3.
T cells and NK cells regulate other immune cells and distinguish between healthy and infected cells.
B cells produce antibodies, which recognize and “tag” foreign invaders (antigens) for a quick immune response if encountered again.
Mucosal tissues act as barriers to pathogens, with antibodies like IgA neutralizing them by preventing receptor binding and triggering enzymes and reactive oxygen species to destroy threats. Lymphocytes and plasma cells collaborate with other white blood cells to defend against pathogens, allergens, and cancer cells, preventing their spread and damage.
Figure 2: In mucosal tissues like the small intestine, memory cells label pathogens as antigens. Dendritic cells transport these antigens to lymph nodes, triggering an immune response.
Abnormal lymphocyte counts can cause symptoms that complicate diagnosis and treatment. Cancer and autoimmune disease drugs may raise lymphocyte levels, risking severe complications.
Figure 3: Chronic abnormal lymphocyte activity is linked to excess proinflammatory proteins and reactive oxygen species (ROS), causing inflammation and damaging organ systems. This dysregulated immune response can harm cells, mutate genes, impair metabolism, and reduce antioxidant activity, contributing to cardiovascular, digestive, and neurological diseases across all ages.
Lymphocyte Formation and Regulation
All blood cells develop from hematopoietic stem cells (HSCs), which differentiate into platelets, reticulocytes (immature RBCs), and leukocytes (WBCs). WBCs form myeloid progenitor cells (CMPs) and lymphoid progenitor cells (CLPs). CMPs produce eosinophils, basophils, monocytes, and neutrophils, while CLPs form T cells, B cells, and natural killer cells. Most WBCs (60-70%) are made in bone marrow, with the rest formed in the thymus, spleen, liver, and lymph nodes.
Figure 4: All blood cells develop from hematopoietic stem cells (hemocytoblasts), which differentiate into myeloid progenitor cells5. Bone marrow, primarily in flat bones like the skull, ribs, pelvis, and sternum, produces immature red blood cells, platelets, and WBCs, which further develop into monocytes, lymphocytes, eosinophils, neutrophils, and basophils.
Lymphocytes, also called mononuclear leukocytes, lack granules and have non-lobular nuclei. Their production in the bone marrow and thymus is regulated by co-stimulatory signals and proinflammatory proteins, such as IL-7 for T cells and IL-3 for B cells.
Immature T cells leave the bone marrow and migrate to the thymus, where they differentiate into gamma-delta or alpha-beta T cells3. Gamma-delta T cells move to tissues, while alpha-beta T cells undergo selection: cells recognizing self-molecules appropriately survive (positive selection), and those reacting too strongly are eliminated (negative selection). Mature alpha-beta T cells express CD4 or CD8 markers and complete their development in the thymus.
B cells leave the bone marrow and mature in the spleen, receiving signals like BAFF to rearrange immunoglobulin (Ig) genes and express IgM as B-cell receptors (BCRs). Upon encountering antigens, T cells become effector or memory cells, and B cells differentiate into plasma cells to produce antibodies. TCR and BCR activation drives lymphocyte proliferation and differentiation.
To prevent excessive activation, mechanisms like the Fas receptor-Fas ligand interaction and regulatory T cells (Tregs) induce programmed lymphocyte death. Cytokines such as IL-2 support survival, while IL-10, TGF-beta, PD-1, and CTLA-4 inhibit overactivation. Balancing lymphocyte production and elimination is crucial to resolving inflammation and preventing autoimmune disorders, infections, cancer, and immunodeficiency.
Clinical Significance of a High Lymphocyte Count (Lymphocytosis)
Lymphocytosis, a high lymphocyte count exceeding 40% of total white blood cells,is linked to chronic bacterial and viral infections (e.g., pertussis, syphilis, staphylococcus), allergies, autoimmune disorders, bone marrow disorders (like leukemia), and cancers (like lymphoma).
Lymphocytosis is linked to dehydration and atherosclerosis, which restrict blood flow (vasoconstriction). Reduced blood flow to the digestive tract lowers stomach acid (HCl) and disrupts gut microbiota, affecting digestion, nutrient absorption, and waste removal. Low HCl and gut imbalance lead to undigested food particles in the gut and blood, increasing blood viscosity and causing oxygen delivery issues (hypoxia). Hypoxia impairs cell function, increases reactive oxygen species (ROS), and causes oxidative stress, damaging cells and mutating genes.
Figure 5: A healthy gut microbiota supports digestion, nutrient absorption, and waste removal, maintaining metabolic balance. Dysbiosis, or reduced healthy bacteria, disrupts metabolism and allows harmful pathogens to influence immune responses.
Oxidative stress occurs when oxygen not bound to hemoglobin oxidizes low-density lipoprotein (LDL) cholesterol, a key membrane component. Oxidized LDL deposits in injured blood vessels, triggering an inflammatory response. The reticuloendothelial system releases immune cells to clear debris, prevent bleeding, and repair tissue. White blood cells break down cholesterol buildup, while platelets and smooth muscle form clots, scar tissue, and new blood vessels, leading to oxidative stress-induced atherosclerosis.
Figure 6: Atherosclerosis thickens vessels, restricts blood flow, raises pressure, and causes organ blood backup. Persistent hypoxia triggers inflammation, damaging organ structure and function 10 .
A 2017 study in the American Journal of Epidemiology linked rising lymphocyte counts to atherosclerosis progression and shorter lifespan 10 . ROS damages genes, hinders cell function, and triggers inflammation, which lymphocytes work to control.
Infections
Lymphocytosis often occurs during infections as lymphocytes combat pathogens like viruses, parasites, fungi, and bacteria, preventing the release of harmful toxins that damage cells. For example, in parasitic infections, T cells recruit eosinophils to release reactive oxygen species (ROS) and cytokines, clearing debris and repairing wounds. However, parasites like helminths can evade immune responses, persisting for years.
Chronic inflammation damages microbiota, restricts blood flow, and activates dormant pathogens like viruses. Viruses, parasitic in nature, replicate by hijacking host cells and releasing toxins, damaging cell membranes and stealing nutrients. A 1994 study found 78% of chronic fatigue syndrome cases had markers of chronic viral infections , which can alter brain structure and function, contributing to mood disorders, dementia, and cognitive decline 16 .
Figure 7: Viruses infect cells by attaching to specific receptors on the host cell surface. They then inject their genetic material or enter through endocytosis. Inside, they hijack the host cell to replicate their genetic material and produce new viral particles, which go on to infect neighboring cells, perpetuating the infection cycle18.
Memory lymphocytes play a key role in quickly responding to recurrent viral infections by producing antibodies and activating cytotoxic responses to eliminate pathogens. However, during chronic inflammation, recycling systems in organs like the liver, intestine, spleen, and kidneys become overwhelmed with toxic byproducts. This diverts lymphocytes from fighting pathogens, allowing infections to persist.
Cancer
Studies suggest that one in ten cancers stems from chronic vascular inflammation and dysbiosis producing reactive oxygen species (ROS), which increase mutated “cancer” cells and support chronic viral infections like HIV, HSV, and EBV. EBV, the first known cancer-causing virus, infects over 90% of people worldwide, often asymptomatically in childhood. Stress can reactivate EBV, leading to conditions like infectious mononucleosis, autoimmune disorders, and cancers such as Hodgkin’s lymphoma and gastric carcinoma. 27 Without addressing stressors, T and B cell dysregulation exacerbates chronic inflammation.
Chronic lymphocytic leukemia (CLL), the most common leukemia worldwide, involves the buildup of abnormal B-lymphocytes and smudge cells in the blood, bone marrow, and lymphoid tissues. It is often detected incidentally during routine blood tests, primarily in older adults, with a median diagnosis age of 7429.
Figure 8: Chronic inflammation increases reactive oxygen species (ROS), proinflammatory proteins, and oxidized cholesterol, activating leukocytes and platelets. This leads to tissue injury, clots, plaques, restricted blood flow, high blood pressure, and venous blood redistribution, driving atherosclerosis, autoimmune diseases, infections, cancer, and organ damage.
Figure 9: Excess oxidized cholesterol, proinflammatory proteins, and reactive oxygen species damage tissues, mutate genes, and reduce cell division. Decreased nitric oxide weakens blood vessels, while oxidized cholesterol embeds in membranes, triggering neutrophil recruitment. Tumor cells exploit this by binding to arterial walls, evading natural killer cells, activating platelets for clots and angiogenesis, and using proinflammatory proteins to grow and invade blood vessels.
Clinical Significance of Low Lymphocyte Count (Lymphopenia)
Lymphopenia, a low lymphocyte count below 20% of total WBCs,weakens immune defense, increasing vulnerability to infections and inflammation-related organ failure . A 2024 study found lymphopenia is more common with age, peaking at 6.84% in people 75 and older, and affecting 41% of hospitalized patients. It is also linked to autoimmune diseases, dysbiosis, dehydration, and vasoconstriction, which impair digestion, nutrient absorption, protein synthesis, and waste excretion. These factors divert immune cells from fighting pathogens to repairing tissues, reducing lymphocyte production.
Dysregulated lymphocytes may attack healthy tissues, triggering autoimmune diseases like diabetes, non-alcoholic fatty liver disease (NAFLD), and cardiovascular diseases, all marked by chronic inflammation.10 Over 90% of the 415 million adults with diabetes in 2015 had type 2 diabetes, with 55% also having NAFLD10. These conditions share a connection to abnormal lymphocyte function and low-grade inflammation.
Type 1 Diabetes Mellitus
Type 1 diabetes mellitus (T1DM) is a chronic autoimmune disease affecting 15 per 100,000 people in the U.S. and 9.5% globally. In 2021, 8.4 million people worldwide had T1DM, with 18% under 20, 64% aged 20–59, and 19% aged 60 or older. That year saw 500,000 new cases, a median onset age of 29, and 35,000 deaths within a year of symptomatic onset. About 1.8 million people with T1DM live in low- and lower-middle-income countries.
In T1DM, persistent hypoxia reduces energy production (ATP) and increases reactive oxygen species (ROS), causing cell damage in organs like the pancreas. The loss of pancreatic cells decreases insulin, glucagon, and leptin production, disrupting glucose uptake and raising blood glucose and ROS levels. This leads to thickened blood, reduced oxygen delivery, and inflammation 10 . Lymphocytes, particularly CD8+ T cells and memory T cells, infiltrate the pancreas, driving autoimmune destruction of beta cells and contributing to oxidative stress and vascular inflammation.
Rheumatoid Arthritis and Systemic Lupus Erythematosus
Lymphopenia is common in autoimmune diseases like rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE).32 In 2020, 17.6 million people globally had RA, a 14.1% increase since 1990, with women affected 2.45 times more than men. RA caused 38,300 deaths and 3.06 million disability-adjusted life years (DALYs), 76.4% from years lived with disability. Smoking accounted for 7.1% of RA-related DALYs. While RA mortality has decreased, its prevalence is projected to reach 31.7 million by 2050, highlighting its growing global burden.
Inflammatory Bowel Diseases
Inflammatory bowel diseases (IBD), including Crohn’s, ulcerative colitis (UC), celiac disease, and types of colitis, are autoimmune disorders affecting the digestive tract. UC is limited to the colon, while Crohn’s and other colitis can impact any part of the tract. Celiac disease, triggered by gluten, primarily affects the small intestine. IBD incidence is 10.9 per 100,000 person-years, with 2.39 million U.S. cases in 2020, mostly among Caucasians and those of Mexican descent.
IBD arises from factors like dysbiosis (SIBO), stress, reduced gut blood flow, and environmental triggers, leading to undigested food particles in the gut that cause inflammation10 . Excess lymphocyte activity, a hallmark of IBD, recruits immune cells to protect intestinal integrity but can damage the gut barrier . This “leaky gut” allows toxins to enter the bloodstream, triggering inflammation, damaging organs, and disrupting metabolic pathways. Elevated enzymes in IBD patients indicate chronic inflammation and impaired healing.
Chronic Obstructive Lung Diseases
In 2021, 14.2 million U.S. adults (6.5%) were diagnosed with chronic lung diseases like COPD, with cases remaining steady since 2011 . COPD is more common in rural areas, linked to higher smoking rates, aging populations, and socioeconomic challenges. About 25% of COPD patients (3.8 million) never smoked.
COPD increases lymphocytes, particularly CD8+ T cells, due to oxidative stress and smoking-related damage, which trigger inflammation and recruit immune cells. Lung inflammation impairs oxygen delivery (hypoxia), damages cells, and promotes platelet production in the lungs,10 where megakaryocytes (MKs) contribute to chronic inflammation. Elevated ROS levels link COPD to atherosclerosis and autoimmune disorders60.
Figure 9: Low blood oxygen boosts red blood cell production and allows megakaryocytes (MKs) to leave the bone marrow and reach the lungs. When oxygen levels rise, lung MKs convert to platelets, which are quickly destroyed. Chronic inflammation accelerates platelet breakdown, reducing liver thrombopoietin production and suppressing bone marrow platelet formation.
Conclusion
Lymphocyte count is a key marker of immune health, reflecting a person’s risk for various conditions. High lymphocyte counts (lymphocytosis) can increase proinflammatory proteins and reactive oxygen species (ROS), damaging cells, mutating genes, and disrupting metabolic pathways. Low lymphocyte counts (lymphopenia) are linked to chronic inflammation, impaired organ function, and a higher risk of diseases like diabetes, IBD, rheumatoid arthritis, COPD, infections, and cancers. Abnormal lymphocyte counts often result from poor nutrition, sleep disturbances, and stress, which affect blood flow, gut health, digestion, nutrient absorption, and hormonal balance.
Managing lymphocyte imbalances requires lifestyle changes, including a healthy diet, proper hydration, regular exercise, and stress reduction. These steps may address the issue without medications, which can sometimes complicate lymphocyte production. A multidisciplinary approach is essential to restore lymphocyte function, improve overall health, and lower the risk of severe conditions.
Source References and Supplemental Research:
Young B, Lowe jo, Stevens A, Heath JW (2006). Wheater’s Functional Histology (5th ed.). Elsevier Limited. ISBN 978-0-443-06850-8.
Dean L. Blood Groups and Red Cell Antigens [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2005. Chapter 1, Blood and the cells it contains. Available from: [PubMed]
LaRosa D, Orange J. 1. lymphocytes. J Allergy Clin Immunol Pract. 2008;121(2). doi:10.1016/j.jaci.2007.06.016 [Crosslink]
The Immune Response against Pathogens. Open Oregon State. Chapter 21-5. [Open Oregon State]
Grinenko T, Eugster A, Thielecke L, et al. Hematopoietic stem cells can differentiate into restricted myeloid progenitors before cell division in mice. Nat Commun. 2018;9(1):1898. Published 2018 May 15. doi:10.1038/s41467-018-04188-7 [PMC Full Text] [PubMed] [Crosslink]
MacNee W. Pathology, pathogenesis, and pathophysiology. BMJ 2006;332:1202. 18 May 2006. doi: https://doi.org/10.1136/bmj.332.7551.1202. [BMJ]
Tahir N, Zahra F. Neutrophilia. [Updated 2023 Apr 27]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan-. Available from: [PubMed]
What Is Gut Dysbiosis? Cleveland Clinic. [Cleveland Clinic]
Hansson GK, Hermansson A. The immune system in atherosclerosis. Nat Immunol. 2011;12:204–12. [PubMed] [Crosslink].
Herrero-Cervera, A., Soehnlein, O. & Kenne, E. Neutrophils in chronic inflammatory diseases. Cell Mol Immunol. 2022;19(2):177-191. doi:10.1038/s41423-021-00832-3 [PubMed] [PMC Full Text] [Crosslink]
Nordestgaard, B. G., & Varbo, A. (2014). Triglycerides and cardiovascular disease. The Lancet, 384(9943), 626–635. https://doi.org/10.1016/s0140-6736(14)61177-6 [Science Direct] [Scopus] [Google Scholar]
Varbo, A., Benn, M., Tybjærg-Hansen, A., & Nordestgaard, B. G. (2013). Elevated remnant cholesterol causes both Low-Grade inflammation and ischemic heart disease, whereas elevated Low-Density lipoprotein cholesterol causes ischemic heart disease without inflammation. Circulation, 128(12), 1298–1309. https://doi.org/10.1161/circulationaha.113.003008 [Scopus] [Google Scholar]
Silvestre-Roig C, Braster Q, Ortega-Gomez A, Soehnlein O. Neutrophils as regulators of cardiovascular inflammation. Nat Rev Cardiol. 2020;17:327–40. [PubMed] [Crosslink].
Soehnlein O, Steffens S, Hidalgo A, Weber C. Neutrophils as protagonists and targets in chronic inflammation. Nat Rev Immunol. 2017;17:248–61. [PubMed] [Crosslink].
Soehnlein O, Libby P. Targeting inflammation in atherosclerosis — from experimental insights to the clinic. Nat Rev Drug Discov. 2021;20:589–610. [PubMed].
Kabat GC, Kim MY, Manson JE, et al. White Blood Cell Count and Total and Cause-Specific Mortality in the Women’s Health Initiative. Am J Epidemiol. 2017;186(1):63-72. doi:10.1093/aje/kww226. [PubMed]
Shin MH, Lee YA, Min DY. Eosinophil-mediated tissue inflammatory responses in helminth infection. Korean J Parasitol. 2009;47 Suppl(Suppl):S125-S131. doi:10.3347/kjp.2009.47.S.S125 [PubMed] [PMC Full Text] [Crosslink]
Alberts B, Johnson A, Lewis J, et al. Molecular Biology of the Cell. 4th edition. New York: Garland Science; 2002. Cell Biology of Infection. [PubMed]
Vandenesch, F, Lina G, Henry T. Staphylococcus aureus hemolysins, bi-component leukocidins, and cytolytic peptides: a redundant arsenal of membrane-damaging virulence factors? Front. Cell. Infect. Microbiol., 15 February 2012. Sec. Bacteria and Host. Volume 2 – 2012. https://doi.org/10.3389/fcimb.2012.00012. [Frontiers]
Hilgers A, Frank J. Chronic Fatigue Syndrome: immune dysfunction, role of pathogens and toxic agents and neurological and cardiac changes. Wien Med Wochenschr. 1994;144(16):399-406. [Pubmed]
Wang Y, Ding L, Zhu Q, Shu M, Cai Q. Common Infections May Lead to Alzheimer’s Disease. Virol Sin. 2018;33(5):456-458. doi:10.1007/s12250-018-0049-7 [Pubmed] [Springer]
Pyzik A, Grywalska E, Matyjaszek-Matuszek B, et al. Does the Epstein-Barr Virus Play a Role in the Pathogenesis of Graves’ Disease?. Int J Mol Sci. 2019;20(13):3145. Published 2019 Jun 27. doi:10.3390/ijms20133145 [Pubmed] [MDPI] [PMC]
Godkin A, Smith KA. Chronic infections with viruses or parasites: breaking bad to make good. Immunology. 2017;150(4):389-396. doi:10.1111/imm.12703 [Pubmed] [Crossref]
Fol M, Karpik W, Zablotni A, Kulesza J, Kulesza E, Godkowicz M, Druszczynska M. Innate Lymphoid Cells and Their Role in the Immune Response to Infections. Cells. 2024; 13(4):335. https://doi.org/10.3390/cells13040335. [MDPI]
Alizon S, Bravo IG, Farrell PJ, Roberts S. Towards a multi-level and a multi-disciplinary approach to DNA oncovirus virulence. Philos Trans R Soc Lond B Biol Sci. 2019;374(1773):20190041. doi:10.1098/rstb.2019.0041 [Pubmed] [The Royal Society] [PMC]
Yu H, Robertson ES. Epstein-Barr Virus History and Pathogenesis. Viruses. 2023;15(3):714. Published 2023 Mar 9. doi:10.3390/v15030714 [PubMed] [PMC Full Text] [Crosslink]
Smatti MK, Al-Sadeq DW, Ali NH, Pintus G, Abou-Saleh H, Nasrallah GK. Epstein-Barr Virus Epidemiology, Serology, and Genetic Variability of LMP-1 Oncogene Among Healthy Population: An Update. Front Oncol. 2018;8:211. Published 2018 Jun 13. doi:10.3389/fonc.2018.00211 [PubMed] [PMC Full Text] [Crosslink]
Lin J, Chen X, Wu H, Chen X, Hu X, Xu J. Peripheral blood lymphocyte counts in patients with infectious mononucleosis or chronic active Epstein-Barr virus infection and prognostic risk factors of chronic active Epstein-Barr virus infection. Am J Transl Res. 2021;13(11):12797-12806. Published 2021 Nov 15. [PubMed] [PMC Full Text]
Milne K, Sturrock B, Chevassut T. Chronic Lymphocytic Leukaemia in 2020: the Future Has Arrived. Curr Oncol Rep. 2020;22(4):36. Published 2020 Mar 14. doi:10.1007/s11912-020-0893-0 [PubMed] [PMC Full Text] [Crosslink]
Kipps TJ, Stevenson FK, Wu CJ, et al. Chronic lymphocytic leukaemia. Nat Rev Dis Primers. 2017;3:16096. Published 2017 Jan 19. doi:10.1038/nrdp.2016.96 [PubMed] [PMC Full Text] [Crosslink]
Dale D. Lymphocytopenia. Merck Manual. April 2023. [Merk Manuals]
Subesinghe S, Kleymann A, Rutherford AI, Bechman K, Norton S, Benjamin Galloway J. The association between lymphopenia and serious infection risk in rheumatoid arthritis. Rheumatology. 2019;59(4):762-766. doi:https://doi.org/10.1093/rheumatology/kez349 [Oxford Academic]
Xie W, Li Q, Ji L, Kang L, Mei J. Prevalence of lymphopenia in the American population: Insights from demographic, BMI, and lifestyle factors. Published online February 12, 2024. doi:10.21203/rs.3.rs-3917749/v1 [Crosslink]
Toori KU, Qureshi MA, Chaudhry A. Lymphopenia: A useful predictor of COVID-19 disease severity and mortality. Pak J Med Sci. 2021;37(7):1984-1988. doi:10.12669/pjms.37.7.4085 [PubMed] [PMC Full Text] [Crosslink]
Wang L, Wang FS, Gershwin ME. Human autoimmune diseases: a comprehensive update. J Intern Med. 2015;278:369–95. [PubMed] [Crosslink].
Sebastian, S., Lucey, B.C. (2008). Reticuloendothelium Malignancy: Current Role of Imaging. In: Blake, M.A., Kalra, M.K. (eds) Imaging in Oncology. Cancer Treatment and Research, vol 143. Springer, Boston, MA. https://doi.org/10.1007/978-0-387-75587-8_17. [SpringerLink]
Dasagrandhi, D., Muthuswamy, A. & Swaminathan, J.K. Atherosclerosis: nexus of vascular dynamics and cellular cross talks. Mol Cell Biochem 477, 571–584 (2022). https://doi.org/10.1007/s11010-021-04307-x. [SpringerLink]
Mandy O J Grootaert, Martin R Bennett, Vascular smooth muscle cells in atherosclerosis: time for a re-assessment, Cardiovascular Research, Volume 117, Issue 11, 1 October 2021, Pages 2326–2339, https://doi.org/10.1093/cvr/cvab046. [Oxford Academic]
Zidar DA, Al-Kindi SG, Liu Y, et al. Association of Lymphopenia With Risk of Mortality Among Adults in the US General Population. JAMA Netw Open. 2019;2(12):e1916526. Published 2019 Dec 2. doi:10.1001/jamanetworkopen.2019.16526 [PubMed] [PMC Full Text] [Crosslink]
Zheng Y, Ley SH, Hu FB. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol. 2018;14:88–98. [PubMed] [Crosslink].
Poznyak A, Grechko AV, Poggio P, Myasoedova VA, Alfieri V, Orekhov AN. The diabetes mellitus–atherosclerosis connection: the role of lipid and glucose metabolism and chronic inflammation. Int J Mol Sci. 2020;21:1835. [PMC Full Text] [Crosslink].
Mobasseri M, Shirmohammadi M, Amiri T, Vahed N, Hosseini Fard H, Ghojazadeh M. Prevalence and incidence of type 1 diabetes in the world: a systematic review and meta-analysis. Health Promot Perspect. 2020;10(2):98-115. Published 2020 Mar 30. doi:10.34172/hpp.2020.18 [PubMed] [PMC Full Text] [Crosslink]
Gregory GA, Robinson TIG, Linklater SE, et al. Global incidence, prevalence, and mortality of type 1 diabetes in 2021 with projection to 2040: a modeling study [published correction appears in Lancet Diabetes Endocrinol. 2022 Nov;10(11):e11. doi: 10.1016/S2213-8587(22)00280-7]. Lancet Diabetes Endocrinol. 2022;10(10):741-760. doi:10.1016/S2213-8587(22)00218-2 [PubMed] [Crosslink]
Valle A, Giamporcaro GM, Scavini M, Stabilini A, Grogan P, Bianconi E, et al. Reduction of circulating neutrophils precedes and accompanies type 1 diabetes. Diabetes. 2013;62:2072–7. [PubMed] [PMC Full Text] [Crosslink].
Vecchio F, Lo Buono N, Stabilini A, Nigi L, Dufort MJ, Geyer S, et al. Abnormal neutrophil signature in the blood and pancreas of presymptomatic and symptomatic type 1 diabetes. JCI Insight. 2018;3:e122146. [PMC Full Text].
Qin J, Fu S, Speake C, Greenbaum CJ, Odegard JM. NETosis-associated serum biomarkers are reduced in type 1 diabetes in association with neutrophil count. Clin Exp Immunol. 2016;184:318–22. [PubMed] [PMC Full Text] [Crosslink].
Dufort MJ, Greenbaum CJ, Speake C, Linsley PS. Cell type-specific immune phenotypes predict loss of insulin secretion in new-onset type 1 diabetes. JCI insight. 2019;4:e125556. [PMC Full Text] [Crosslink].
M Radenkovic, K Uvebrant, O Skog, L Sarmiento, J Avartsson, P Storm, P Vickman, P-A Bertilsson, M Fex, O Korgsgren, C M Cilio, Characterization of resident lymphocytes in human pancreatic islets, Clinical and Experimental Immunology, Volume 187, Issue 3, March 2017, Pages 418–427, https://doi.org/10.1111/cei.12892. [Oxford Academic]
Sun, F., Yang, CL., Wang, FX. et al. Pancreatic draining lymph nodes (PLNs) serve as a pathogenic hub contributing to the development of type 1 diabetes. Cell Biosci 13, 156 (2023). https://doi.org/10.1186/s13578-023-01110-7. [BioMed Central]
GBD 2021 Rheumatoid Arthritis Collaborators. Global, regional, and national burden of rheumatoid arthritis, 1990-2020, and projections to 2050: a systematic analysis of the Global Burden of Disease Study 2021. Lancet Rheumatol. 2023;5(10):e594-e610. Published 2023 Sep 25. doi:10.1016/S2665-9913(23)00211-4 [PubMed] [PMC Full Text] [Crosslink]
McDowell C, Farooq U, Haseeb M. Inflammatory Bowel Disease. [Updated 2023 Aug 4]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK470312/
Lewis JD, Parlett LE, Jonsson Funk ML, et al. Incidence, Prevalence, and Racial and Ethnic Distribution of Inflammatory Bowel Disease in the United States. Gastroenterology. 2023;165(5):1197-1205.e2. doi:10.1053/j.gastro.2023.07.003 [PubMed] [Crosslink]
Zhang YZ, Li YY. Inflammatory bowel disease: pathogenesis. World J Gastroenterol 2014;20:91–9. [PubMed] [PMC Full Text] [Crosslink].
Gomez-Bris R, Saez A, Herrero-Fernandez B, Rius C, Sanchez-Martinez H, Gonzalez-Granado JM. CD4 T-Cell Subsets and the Pathophysiology of Inflammatory Bowel Disease. International Journal of Molecular Sciences. 2023; 24(3):2696. https://doi.org/10.3390/ijms24032696. [MDPI]
Nikolakis D, de Voogd FAE, Pruijt MJ, Grootjans J, van de Sande MG, D’Haens GR. The Role of the Lymphatic System in the Pathogenesis and Treatment of Inflammatory Bowel Disease. International Journal of Molecular Sciences. 2022; 23(3):1854. https://doi.org/10.3390/ijms23031854. [MDPI]
Liu Y, Carlson SA, Watson KB, Xu F, Greenlund KJ. Trends in the Prevalence of Chronic Obstructive Pulmonary Disease Among Adults Aged ≥18 Years — United States, 2011–2021. MMWR Morb Mortal Wkly Rep 2023;72:1250–1256. DOI: http://dx.doi.org/10.15585/mmwr.mm7246a1.
Williams, M., Todd, I. & Fairclough, L.C. The role of CD8 + T lymphocytes in chronic obstructive pulmonary disease: a systematic review. Inflamm. Res. 70, 11–18 (2021). https://doi.org/10.1007/s00011-020-01408-z. [SpringerLink]
Zhao. Platelet generation from circulating megakaryocytes is triggered in the lung vasculature. BioRXiv (2021) bioRxiv 2021.11.01.466743. doi: 10.21203/rs.3.rs-690639/v1 [BioRXiv Full Text] [Crosslink]
Gelon L, Fromont L, Lefrançais E. Occurrence and role of lung megakaryocytes in infection and inflammation. Frontiers in Immunology. 2022;13. doi:10.3389/fimmu.2022.1029223 [PubMed] [PMC Full Text] [Crosslink]
Lefrançais, E., Ortiz-Muñoz, G., Caudrillier, A. et al. The lung is a site of platelet biogenesis and a reservoir for haematopoietic progenitors. Nature 544, 105–109 (2017). https://doi.org/10.1038/nature21706. [Nature].[NIH]
Wirth MD, Sevoyan M, Hofseth L, Shivappa N, Hurley TG, Hébert JR. The Dietary Inflammatory Index is associated with elevated white blood cell counts in the National Health and Nutrition Examination Survey. Brain Behav Immun. 2018;69:296-303. doi:10.1016/j.bbi.2017.12.003 [PubMed] [PMC Full Text] [Crosslink]
Cedars-Sinai Staff. Are animal proteins better for you than plant proteins? Cedars Sinai. January 16, 2019. Accessed September 2, 2024. [Website]
Koenig G, Seneff S. Gamma-glutamyltransferase: A predictive biomarker of cellular antioxidant inadequacy and disease risk. Dis Markers. 2015:2015:818570. PMID: 26543300. doi: 10.1155/2015/818570. [PubMed] [Wiley] [PubMed]
De Chiara, F., Checcllo, C. U., & Azcón, J. R. (2019). High protein diet and metabolic plasticity in Non-Alcoholic Fatty liver Disease: Myths and Truths. Nutrients, 11(12), 2985. https://doi.org/10.3390/nu11122985 [PubMed]
Balkaransingh PD, Wheeler D, Ning Y, Helou MA, Massey G. Dietary determinants of the White Blood Cell Count. Blood. 2013;122(21):1705-1705. doi:10.1182/blood.v122.21.1705.1705 [Crosslink] [Full Text]
Guoyao Wu; Yun-Zhong Fang; Sheng Yang; Joanne R. Lupton; Nancy D. Turner (2004). “Glutathione Metabolism and its Implications for Health”. Journal of Nutrition. 134 (3): 489–492. doi:10.1093/jn/134.3.489. PMID 14988435 [PubMed] [Crosslink]
Ho, P.I.; Ortiz, D.; Rogers, E.; Shea, T.B. Multiple aspects of homocysteine neurotoxicity: Glutamate excitotoxicity, kinase hyperactivation and DNA damage. J. Neurosci. Res. 2002, 70, 694–702. [Google Scholar] [CrossRef] [PubMed]
Menni C, Louca P, Berry SE, et al. High intake of vegetables is linked to lower white blood cell profile and the effect is mediated by the gut microbiome. BMC Med. 2021;19(1):37. Published 2021 Feb 11. doi:10.1186/s12916-021-01913-w [PubMed] [PMC Full Text] [Crosslink]
Hernáez Á, Lassale C, Castro-Barquero S, Babio N, Ros E, Castañer O, Tresserra-Rimbau A, Pintó X, Martínez-González MÁ, Corella D, et al. Mediterranean Diet and White Blood Cell Count—A Randomized Controlled Trial. Foods. 2021; 10(6):1268. https://doi.org/10.3390/foods10061268. [MDPII]
Moll S, Varga EA. Homocysteine and MTHFR Mutations. Circulation. 2015;132(1). doi:https://doi.org/10.1161/circulationaha.114.013311 [AHA Journals] [Crosslink]
Esse, R.; Barroso, M.; Almeida, I.; Castro, R. The contribution of homocysteine metabolism disruption to endothelial dysfunction: State-of-the-art. Int. J. Mol. Sci. 2019, 20, 867. [Google Scholar] [CrossRef] [Green Version].
Kruman, I.I.; Culmsee, C.; Chan, S.L.; Kruman, Y.; Guo, Z.; Penix, L.; Mattson, M.P. Homocysteine elicits a DNA damage response in neurons that promotes apoptosis and hypersensitivity to excitotoxicity. J. Neurosci. 2000, 20, 6920–6926. [PubMed]
Ho, P.I.; Ashline, D.; Dhitavat, S.; Ortiz, D.; Collins, S.C.; Shea, T.B.; Rogers, E. Folate deprivation induces neurodegeneration: Roles of oxidative stress and increased homocysteine. Neurobiol. Dis. 2003, 14, 32–42. [PubMed]
Piskin E, Cianciosi D, Gulec S, Tomas M, Capanoglu E. Iron Absorption: Factors, Limitations, and Improvement Methods. ACS Omega. 2022 Jun 21; 7(24): 20441–20456. doi: 10.1021/acsomega.2c01833 [PMC] [Crosslink]
Boldt D. H. New Perspectives on Iron: An Introduction. Am. J. Med. Sci. 1999, 318 (4), 207. 10.1097/00000441-199910000-00001. [PubMed]
Smirnoff N. Ascorbic Acid Metabolism and Functions: A Comparison of Plants and Mammals. Free Radic. Biol. Med. 2018, 122, 116–129. 10.1016/j.freeradbiomed.2018.03.033. [PubMed]
Benkhedda K.; L’abbé M. R.; Cockell K. A. Effect of Calcium on Iron Absorption in Women with Marginal Iron Status. Br. J. Nutr. 2010, 103 (5), 742–748. 10.1017/S0007114509992418. [PubMed] [Cambridge Core]
Ziegler E.E. Consumption of cow’s milk as a cause of iron deficiency in infants and toddlers. Nutr. Rev. 2011;69:37–42. doi: 10.1111/j.1753-4887.2011.00431.x. [PubMed] [Oxford Academic]
Cory H, Passarelli S, Szeto J, Tamez M, Mattei J. The Role of Polyphenols in Human Health and Food Systems: A Mini-Review. Front Nutr. 2018;5:87. Published 2018 Sep 21. doi:10.3389/fnut.2018.00087 [PubMed]
Zhang H, Tsao R. Dietary polyphenols, oxidative stress and antioxidant and anti-inflammatory effects. Curr Opin Food Sci. (2016) 8:33–42. 10.1016/j.cofs.2016.02.002 [CrossRef] [Google Scholar]
Sandberg A-S, Scheers N. Phytic Acid: Properties, Uses, and Determinations. In: ; :365-368. Accessed September 2, 2024. [ScienceDirect]
Hart J. J.; Tako E.; Kochian L. V.; Glahn R. P. Identification of Black Bean (Phaseolus Vulgaris L.) Polyphenols That Inhibit and Promote Iron Uptake by Caco-2 Cells. J. Agric. Food Chem. 2015, 63 (25), 5950–5956. 10.1021/acs.jafc.5b00531. [PubMed]
Naghedi-Baghdar, H., Nematy, M., Kooshyar, M. M., Taghipour, A., Sajadi Tabassi, S. A., Shokri, S., Javan, R., & Nazari, S. M. (2018). Effect of a functional food (vegetable soup) on blood rheology in patients with polycythemia. Avicenna journal of phytomedicine, 8(5), 389–398. [PubMed]
Watso, J. C., & Farquhar, W. B. (2019). Hydration Status and Cardiovascular Function. Nutrients, 11(8), 1866. https://doi.org/10.3390/nu11081866 [PubMed]
Popkin BM, D’Anci KE, Rosenberg IH. Water, hydration, and health. Nutr Rev. 2010;68(8):439-458. doi:10.1111/j.1753-4887.2010.00304.x [PubMed]
Billett HH. Hemoglobin and Hematocrit. In: Walker HK, Hall WD, Hurst JW, editors. Clinical Methods: The History, Physical, and Laboratory Examinations. 3rd edition. Boston: Butterworths; 1990. Chapter 151. Available from: [PubMed]
Kennedy M. How much water you’re actually supposed to drink each day – and why 8 cups isn’t right for everyone. Business Insider. December 14, 2021. [Website]
Pałka T, Koteja PM, Tota Ł, et al. The Influence of Various Hydration Strategies (Isotonic, Water, and No Hydration) on Hematological Indices, Plasma Volume, and Lactate Concentration in Young Men during Prolonged Cycling in Elevated Ambient Temperatures. Biology (Basel). 2023;12(5):687. Published 2023 May 7. doi:10.3390/biology12050687 [PubMed] [PMC Full Text] [Crosslink]
Ballard H. S. (1997). The hematological complications of alcoholism. Alcohol health and research world, 21(1), 42–52. [PubMed]
Bradley CS, Erickson BA, Messersmith EE, et al. Evidence of the Impact of Diet, Fluid Intake, Caffeine, Alcohol and Tobacco on Lower Urinary Tract Symptoms: A Systematic Review. J Urol. 2017;198(5):1010-1020. doi:10.1016/j.juro.2017.04.097 [PubMed] [PMC Full Text] [Full Text]
Polhuis KCMM, Wijnen AHC, Sierksma A, Calame W, Tieland M. The Diuretic Action of Weak and Strong Alcoholic Beverages in Elderly Men: A Randomized Diet-Controlled Crossover Trial. Nutrients. 2017;9(7):660. Published 2017 Jun 28. doi:10.3390/nu9070660 [PubMed]
Chung PS, Tsai KZ, Lin YP, Lin YK, Lin GM. Association between Leukocyte Counts and Physical Fitness in Male Military Members: The CHIEF Study. Sci Rep. 2020;10(1):6082. Published 2020 Apr 8. doi:10.1038/s41598-020-63147-9 [PubMed] [PMC Full Text] [Crosslink]
Johannsen NM, Swift DL, Johnson WD, et al. Effect of different doses of aerobic exercise on total white blood cell (WBC) and WBC subfraction number in postmenopausal women: results from DREW. PLoS One. 2012;7(2):e31319. doi:10.1371/journal.pone.0031319 [PubMed] [PMC Full Text] [Crosslink]
El-Sayed M. S. (1998). Effects of exercise and training on blood rheology. Sports medicine (Auckland, N.Z.), 26(5), 281–292. https://doi.org/10.2165/00007256-199826050-00001 [PubMed] [Springer]
Kilic-Toprak, E., Ardic, F., Erken, G., Unver-Kocak, F., Kucukatay, V., & Bor-Kucukatay, M. (2012). Hemorheological responses to progressive resistance exercise training in healthy young males. Medical science monitor : international medical journal of experimental and clinical research, 18(6), CR351–CR360. https://doi.org/10.12659/msm.882878 [PubMed] [ISI] [PubMed]
Kamal A, Malik RN. Hematological Evidence of Occupational Exposure to Chemicals and Other Factors among Auto-Repair Workers in Rawalpindi, Pakistan. Osong Public Health Res Perspect. 2012;3(4):229-238. doi:10.1016/j.phrp.2012.10.003 [PubMed]
Premont RT, Reynolds JD, Zhang R, Stamler JS. Role of Nitric Oxide Carried by Hemoglobin in Cardiovascular Physiology: Developments on a Three-Gas Respiratory Cycle. Circ Res. 2020;126(1):129-158. doi:10.1161/CIRCRESAHA.119.315626 [PubMed] [PMC Full Text] [Circulation Research]
Pedersen KM, Çolak Y, Ellervik C, Hasselbalch HC, Bojesen SE, Nordestgaard BG. Smoking and Increased White and Red Blood Cells. Arterioscler Thromb Vasc Biol. 2019;39(5):965-977. doi:10.1161/ATVBAHA.118.312338 [PubMed] [Crosslink]
Carel RS, Eviatar J. Factors affecting leukocyte count in healthy adults. Prev Med. 1985;14(5):607-619. doi:10.1016/0091-7435(85)90081-7 [PubMed] [Crosslink]
Pillai AA, Fazal S, Mukkamalla SKR, Babiker HM. Polycythemia. In: StatPearls. Treasure Island (FL): StatPearls Publishing; May 20, 2023. [PubMed]