Type 1 Diabetes: Pathogenesis, Risk Factors, Management, Complications, and the Impact of COVID-19

Comprehensive Review of Type 1 Diabetes Mellitus: Pathogenesis, Management, Complications, and Emerging Insights from the COVID-19 Pandemic

Many thanks to our sponsor Esdebe who helped us prepare this research report.

Abstract

Type 1 diabetes mellitus (T1D) is a multifaceted chronic autoimmune disorder characterized by the progressive, immune-mediated destruction of insulin-producing pancreatic beta cells, ultimately leading to absolute insulin deficiency and chronic hyperglycemia. This extensive report provides an in-depth examination of T1D, delving into its intricate pathogenesis, a broad spectrum of genetic and environmental risk factors, contemporary management paradigms, the full range of acute and chronic complications, and the cutting-edge landscape of research aimed at prevention and curative strategies. Special emphasis is placed on the profound impact of the COVID-19 pandemic on the incidence of new-onset T1D and the implications for clinical practice and long-term patient outcomes, incorporating the latest epidemiological and mechanistic insights.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

1. Introduction

Type 1 diabetes mellitus (T1D), historically known as juvenile diabetes or insulin-dependent diabetes, represents a complex autoimmune condition primarily orchestrated by the adaptive immune system’s erroneous assault on the insulin-secreting beta cells situated within the pancreatic islets of Langerhans. This relentless destruction culminates in an absolute deficiency of insulin, a vital hormone indispensable for glucose uptake by cells, thereby leading to chronic elevation of blood glucose levels, or hyperglycemia [1]. Distinct from type 2 diabetes (T2D), which predominantly involves insulin resistance and a relative insulin deficiency, T1D is characterized by a profound and irreversible deficit in insulin production. The global incidence of T1D has exhibited a consistent upward trend over the past few decades, particularly among younger populations, transforming it into a pressing public health concern and stimulating intensive research into its multifactorial etiology, sophisticated management approaches, and the ambitious pursuit of preventive and curative interventions [2]. The unprecedented emergence of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), responsible for the COVID-19 pandemic, has introduced novel layers of complexity and urgency into the realm of T1D, notably raising profound questions regarding its potential influence on the incidence of new-onset cases and the optimal management of individuals living with established T1D during and beyond the pandemic era.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

2. Pathogenesis of Type 1 Diabetes

T1D is fundamentally an autoimmune disorder, where the body’s meticulously regulated immune system mistakenly identifies the self-antigens expressed by pancreatic beta cells as foreign invaders, instigating a destructive inflammatory response. This targeted immune assault on beta cells leads to their progressive demise, culminating in an absolute insulin deficiency and subsequent hyperglycemia. The precise sequence of events and the underlying triggers that initiate this autoimmune cascade remain subjects of intensive investigation, yet a consensus is emerging regarding the interplay of genetic predisposition, environmental factors, and intrinsic immune system dysregulation [3, 4].

2.1. Genetic Susceptibility

Genetic predisposition constitutes the most significant risk factor for T1D, with the Human Leukocyte Antigen (HLA) complex contributing the largest proportion of genetic risk. Located on chromosome 6, HLA genes encode proteins critical for presenting antigens to T lymphocytes, thereby playing a central role in immune recognition and self-tolerance. Specific HLA class II alleles, particularly HLA-DR3 and HLA-DR4, as well as HLA-DQ2 and HLA-DQ8, are strongly associated with increased susceptibility to T1D. For instance, individuals carrying both HLA-DR3/DR4-DQ8 haplotypes exhibit a significantly elevated risk, often cited as being 20-50 times higher than the general population, primarily due to their propensity to present self-peptides that activate autoreactive T cells [5, 6].

Beyond the HLA locus, over 60 non-HLA genes have been identified through genome-wide association studies (GWAS) as contributing to T1D risk, each typically conferring a modest individual effect but collectively increasing overall susceptibility. Notable non-HLA genes include:

  • PTPN22 (Protein Tyrosine Phosphatase Non-Receptor Type 22): A variant of this gene (R620W) is associated with increased risk of T1D and other autoimmune diseases. It encodes a lymphoid-specific phosphatase that negatively regulates T-cell activation, and the risk allele may lead to hyperactive T cells [7].
  • INS (Insulin Gene) Variable Number Tandem Repeat (VNTR): Located upstream of the insulin gene, certain VNTR alleles influence the thymic expression of insulin. Low-expressing alleles are linked to reduced central tolerance to insulin, allowing autoreactive T cells to escape deletion in the thymus [8].
  • CTLA4 (Cytotoxic T-Lymphocyte-Associated Protein 4): This gene encodes an inhibitory receptor on T cells that downregulates T-cell activation. Polymorphisms in CTLA4 can impair its inhibitory function, leading to unchecked immune responses [9].
  • IFIH1 (Interferon Induced With Helicase C Domain 1): Encodes an innate immune receptor that detects viral RNA. Variants can alter the innate immune response to viral infections, potentially exacerbating beta-cell damage [10].
  • ERBB3 (Erb-B2 Receptor Tyrosine Kinase 3): Involved in cell proliferation and survival, its role in T1D pathogenesis is less clear but points to broader cellular pathways [11].

These genetic variations are thought to influence various immunological processes, including antigen presentation, T-cell activation, B-cell function, and cytokine signaling, ultimately contributing to a breakdown in immunological tolerance and the initiation of beta-cell destruction.

2.2. Environmental Triggers

While genetic predisposition establishes the susceptibility, environmental factors are hypothesized to act as crucial triggers, initiating or accelerating the autoimmune process in genetically vulnerable individuals. The ‘accelerator hypothesis’ suggests that environmental factors increase the rate of beta-cell destruction in genetically predisposed individuals, leading to earlier disease onset [12].

  • Viral Infections: Viral infections have long been considered prime candidates for environmental triggers due to their potential to induce inflammation, cause direct beta-cell damage, or trigger molecular mimicry.

    • Enteroviruses: Coxsackievirus B (CVB) infections, particularly CVB4, are among the most frequently implicated viruses. Proposed mechanisms include direct cytolytic infection of beta cells, leading to their destruction, and bystander activation of immune cells through inflammation [13]. Molecular mimicry, where viral peptides share sequence homology with beta-cell autoantigens, could lead to cross-reactive immune responses.
    • Other viruses, such as rubella virus (especially congenital rubella syndrome), mumps virus, cytomegalovirus (CMV), and Epstein-Barr virus (EBV), have also been investigated, although definitive causal links remain challenging to establish [14].
  • Dietary Factors:

    • Cow’s Milk Exposure: Early exposure to cow’s milk proteins, particularly bovine insulin or bovine serum albumin (BSA), has been debated as a potential trigger, particularly in infants. The hypothesis suggests that these proteins could initiate molecular mimicry, leading to an immune response against human insulin or other beta-cell proteins [15]. However, large prospective studies have yielded conflicting results, making this association inconclusive.
    • Gluten: Some studies have explored a link between gluten consumption and T1D risk, particularly given the high prevalence of celiac disease in T1D patients. While a direct causal link between gluten and T1D development is not definitively established, dietary gluten can influence gut microbiota and immune responses [16].
    • Vitamin D Deficiency: Vitamin D plays a crucial role in immune modulation. Deficiency has been associated with increased risk of several autoimmune diseases, including T1D, possibly by influencing regulatory T cell function and reducing pro-inflammatory cytokine production [17]. However, interventional studies with vitamin D supplementation for T1D prevention have not yet shown consistent positive outcomes.
  • Gut Microbiome Dysbiosis: The composition and function of the gut microbiota are increasingly recognized for their profound influence on immune system development and regulation. Dysbiosis, an imbalance in the gut microbial community, characterized by reduced microbial diversity or altered metabolic products, has been observed in individuals at risk for T1D and in newly diagnosed patients. This dysbiosis may contribute to impaired gut barrier function (‘leaky gut’), increased systemic inflammation, and altered immune responses, potentially facilitating the autoimmune attack on beta cells [18].

  • Other Environmental Factors:

    • Hygiene Hypothesis: This theory suggests that reduced exposure to infections and microbes in early life, due to improved hygiene, alters immune system development, making individuals more susceptible to autoimmune diseases.
    • Toxins: Certain chemical toxins, such as N-nitroso compounds found in cured meats, have been hypothesized to contribute, but evidence is limited.
    • Accelerated Growth/Obesity: Rapid weight gain in infancy or childhood has been linked to increased T1D risk, possibly by imposing metabolic stress on beta cells [12].

2.3. Immune System Dysregulation

The autoimmune destruction of beta cells is a highly coordinated process involving multiple components of both the innate and adaptive immune systems, driven by a breakdown in immunological tolerance.

  • Antigen Presentation: Autoantigens, such as insulin, glutamic acid decarboxylase (GAD65), insulinoma-associated antigen 2 (IA-2), and zinc transporter 8 (ZnT8), are released from stressed or damaged beta cells. These are processed and presented by antigen-presenting cells (APCs), such as dendritic cells (DCs) and macrophages, to T cells in the pancreatic lymph nodes. This presentation occurs in the context of specific HLA molecules, activating autoreactive T lymphocytes [19].

  • T-Lymphocyte Involvement:

    • CD4+ Helper T cells (Th1): Upon activation by APCs, autoreactive CD4+ T cells differentiate into Th1 cells, producing pro-inflammatory cytokines like interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α). These cytokines enhance inflammation within the pancreatic islets (insulitis) and can directly contribute to beta-cell damage [20].
    • CD8+ Cytotoxic T lymphocytes (CTLs): These cells are considered the primary executioners of beta-cell destruction. Activated CD8+ T cells recognize beta cells presenting autoantigens via HLA class I molecules and directly induce beta-cell apoptosis through mechanisms involving perforin and granzymes, or Fas/FasL pathways [21].
  • B-Lymphocyte Role: While T cells are the direct mediators of beta-cell destruction, B lymphocytes play crucial roles as APCs and producers of autoantibodies (e.g., GAD65 autoantibodies, IA-2 autoantibodies, insulin autoantibodies (IAA), ZnT8 autoantibodies). These autoantibodies are diagnostic markers and reflect the autoimmune process but are not believed to be directly pathogenic in beta-cell destruction, though they may contribute through antibody-dependent cellular cytotoxicity (ADCC) or complement activation [22].

  • Breakdown of Immunological Tolerance:

    • Regulatory T cells (Tregs): These specialized T cells are crucial for maintaining self-tolerance by suppressing autoreactive immune responses. In T1D, there is often a quantitative or functional defect in Tregs, impairing their ability to control the autoimmune attack [23].
    • Central Tolerance: Defects in thymic negative selection can lead to the escape of autoreactive T cells into the periphery.
    • Peripheral Tolerance: Insufficient anergy induction or deletion of autoreactive T cells in peripheral tissues further contributes to the breakdown of tolerance.
  • Insulitis: The inflammatory infiltration of pancreatic islets by immune cells (lymphocytes, macrophages) is a hallmark of pre-symptomatic and early T1D. This ‘insulitis’ indicates ongoing beta-cell destruction and is a key pathological feature [24].

2.4. Beta-Cell Stress and Apoptosis

Beyond immune assault, intrinsic beta-cell factors contribute to their vulnerability. Chronic metabolic stress (e.g., from hyperglycemia, insulin resistance), endoplasmic reticulum (ER) stress, and oxidative stress can impair beta-cell function and survival, making them more susceptible to immune-mediated destruction. Pro-inflammatory cytokines (e.g., IL-1β, IFN-γ, TNF-α) secreted by infiltrating immune cells directly induce beta-cell apoptosis, further exacerbating the loss of insulin production capacity [25].

Many thanks to our sponsor Esdebe who helped us prepare this research report.

3. Risk Factors for Type 1 Diabetes

The development of T1D is a complex interplay of multiple risk factors, leading to a varying degree of susceptibility among individuals. While the exact trigger for the autoimmune attack remains elusive, several factors have been consistently identified as increasing the likelihood of disease onset.

3.1. Genetic Predisposition and Family History

As previously discussed, genetic factors are the predominant determinant of T1D risk. A family history of T1D significantly increases an individual’s susceptibility. The risk for first-degree relatives (siblings, children, parents) of an individual with T1D is approximately 5-10%, which is considerably higher than the 0.4% risk in the general population [26]. For identical twins, if one twin develops T1D, the other twin has a concordance rate of approximately 30-50%, illustrating the significant, but not absolute, role of genetics, indicating that environmental factors are also crucial [27].

The presence of high-risk HLA genotypes, particularly DR3/DR4-DQ8 heterozygosity, confers the highest genetic risk. Furthermore, the cumulative effect of multiple non-HLA gene variants, each contributing a small amount of risk, further stratifies an individual’s overall genetic susceptibility. Genetic screening for these risk alleles is increasingly used in research settings to identify individuals at higher risk for T1D, particularly in the context of prevention trials [28].

3.2. Geographical and Ethnic Variations

The incidence of T1D exhibits remarkable geographical variation, with the highest rates observed in Scandinavian countries, particularly Finland and Sweden, and other countries of predominantly European ancestry [29]. For instance, Finland has one of the highest incidences globally, while rates are significantly lower in East Asian countries. This ‘diabetes belt’ in Northern Europe suggests a strong interaction between genetic predisposition (common among European populations) and specific environmental factors prevalent in these regions (e.g., certain viral exposures, dietary patterns, or vitamin D levels). Incidence rates are generally lower in populations of African, Hispanic, and Asian descent, although global incidence is rising across all ethnic groups [30]. These disparities underscore the complex interplay of genetic ancestry, environmental exposures, and socioeconomic determinants in disease prevalence.

3.3. Age and Sex

T1D can manifest at any age, from infancy to late adulthood, but it is most frequently diagnosed in children and adolescents, with two common peaks in incidence: one around 5-7 years of age and another during puberty (10-14 years). This age-related pattern suggests that developmental changes in the immune system, exposure to environmental triggers, or periods of rapid growth and metabolic stress (e.g., during puberty, when insulin demand increases) may influence disease onset [31].

Regarding sex, studies have shown varying patterns. In younger children, there may be a slight male predominance, while some data suggest a slight female predominance in older adolescents or young adults. Overall, the incidence rate is largely similar between sexes, with minor differences potentially reflecting complex interactions between sex hormones, immune development, and environmental exposures [32].

3.4. Environmental Factors (Expanded)

As detailed in the pathogenesis section, various environmental elements are considered risk factors due to their potential to trigger or accelerate the autoimmune process:

  • Viral Infections: Specific enteroviruses (e.g., Coxsackievirus B), rubella, mumps, and CMV are consistently investigated. Early-life exposure, severity of infection, and genetic susceptibility to viral-induced immune responses are key considerations.
  • Dietary Factors: Early infant diet, particularly cow’s milk protein exposure before 3-4 months of age, and gluten introduction, continue to be areas of active research, though definitive evidence is lacking for prevention [15, 16]. Lower levels of Vitamin D intake or sun exposure are also considered potential risk factors [17].
  • Gut Microbiome: Dysbiosis in early life, characterized by reduced diversity or altered bacterial composition, is a significant emerging risk factor, influencing immune maturation and barrier function [18].
  • Hygiene Hypothesis: Reduced exposure to childhood infections or microorganisms, due to improved sanitation and smaller family sizes, may alter immune system programming, leading to increased susceptibility to autoimmunity [33].

3.5. Presence of Autoantibodies

The presence of multiple autoantibodies targeting pancreatic beta-cell antigens serves as a strong predictive marker for T1D development, even before clinical symptoms appear. These autoantibodies include:

  • Insulin Autoantibodies (IAA): Often the first to appear, particularly in young children, and indicate an immune response against insulin itself.
  • Glutamic Acid Decarboxylase Autoantibodies (GADA): Target GAD65, an enzyme involved in neurotransmitter synthesis in beta cells.
  • Insulinoma-Associated Antigen 2 Autoantibodies (IA-2A): Target IA-2, a protein involved in neuroendocrine granule exocytosis.
  • Zinc Transporter 8 Autoantibodies (ZnT8A): Target ZnT8, a protein involved in insulin crystallization within beta cells.

The number of positive autoantibodies correlates with the risk and speed of progression to clinical T1D. Individuals with two or more autoantibodies have an almost 100% lifetime risk of developing T1D, highlighting their utility in identifying individuals in preclinical stages [34]. This understanding has led to the proposed staging of T1D:

  • Stage 1: Presence of two or more autoantibodies with normal glucose tolerance.
  • Stage 2: Presence of two or more autoantibodies with impaired glucose tolerance (dysglycemia).
  • Stage 3: Clinical onset of T1D (symptomatic diabetes).

Many thanks to our sponsor Esdebe who helped us prepare this research report.

4. Clinical Presentation and Diagnosis

The clinical presentation of T1D typically involves a relatively acute onset of symptoms, often driven by profound insulin deficiency. Recognition and prompt diagnosis are critical to prevent life-threatening complications.

4.1. Symptoms

Classic symptoms of T1D are often referred to as the ‘4 Ps’:

  • Polyuria: Increased urination, due to osmotic diuresis caused by high glucose levels in the kidneys.
  • Polydipsia: Increased thirst, a compensatory mechanism for fluid loss through polyuria.
  • Polyphagia: Increased hunger, despite eating, as cells are starved of glucose due to lack of insulin.
  • Weight Loss: Unexplained weight loss, resulting from the body breaking down fat and muscle for energy due to glucose unavailability.

Other common symptoms include profound fatigue, blurred vision (due to osmotic changes in the eye lens), recurrent infections (e.g., candidiasis), and, in severe cases, abdominal pain, nausea, vomiting, and a fruity odor on the breath (signs of diabetic ketoacidosis) [35].

4.2. Diagnostic Criteria

Diagnosis of T1D is based on established biochemical criteria for hyperglycemia, as defined by organizations such as the American Diabetes Association (ADA) and the World Health Organization (WHO) [36]:

  • Fasting Plasma Glucose (FPG): ≥ 126 mg/dL (7.0 mmol/L) on two separate occasions (fasting for at least 8 hours).
  • Oral Glucose Tolerance Test (OGTT): Plasma glucose ≥ 200 mg/dL (11.1 mmol/L) two hours after a 75-gram glucose load.
  • Glycated Hemoglobin (HbA1c): ≥ 6.5% (48 mmol/mol). While primarily used for monitoring, it can be diagnostic in the absence of conditions affecting red blood cell turnover.
  • Random Plasma Glucose: ≥ 200 mg/dL (11.1 mmol/L) in a patient with classic symptoms of hyperglycemia.

In ambiguous cases, or to differentiate T1D from other forms of diabetes, autoantibody testing (GADA, IA-2A, IAA, ZnT8A) is crucial. The presence of at least one of these autoantibodies is indicative of autoimmune beta-cell destruction, supporting a diagnosis of T1D, especially when C-peptide levels (a marker of endogenous insulin production) are low or undetectable [37].

4.3. Differential Diagnosis

Differentiating T1D from other forms of diabetes is critical for appropriate management. Key distinctions include:

  • Type 2 Diabetes (T2D): T2D typically involves insulin resistance and relative insulin deficiency, often presenting in older, overweight individuals, but can occur in children. Absence of autoantibodies and higher C-peptide levels usually distinguish T2D from T1D.
  • Monogenic Diabetes (MODY): Maturity-onset diabetes of the young (MODY) is caused by single gene mutations affecting beta-cell function, often inherited in an autosomal dominant pattern. It can mimic T1D or T2D but is characterized by a lack of autoantibodies and a more stable, often less severe course [38]. Genetic testing is required for definitive diagnosis.
  • Secondary Diabetes: Diabetes can also arise secondary to other conditions (e.g., pancreatitis, cystic fibrosis, certain medications like glucocorticoids). Clinical history and specific investigations help in these cases.

4.4. Diabetic Ketoacidosis (DKA) at Presentation

Due to the rapid and profound nature of beta-cell destruction, a significant proportion of individuals, particularly children, present with diabetic ketoacidosis (DKA) at the time of T1D diagnosis. DKA is a life-threatening complication resulting from severe insulin deficiency, leading to hyperglycemia, dehydration, ketosis, and metabolic acidosis. Its symptoms are more severe and include deep, rapid breathing (Kussmaul respirations), severe abdominal pain, nausea, vomiting, altered mental status, and a fruity breath odor. DKA requires immediate medical attention and intensive care management [39].

Many thanks to our sponsor Esdebe who helped us prepare this research report.

5. Management Strategies for Type 1 Diabetes

Effective management of T1D is a lifelong endeavor that necessitates a comprehensive, individualized, and patient-centered approach. The primary goal is to maintain blood glucose levels within a target range to prevent both acute and chronic complications, thereby optimizing quality of life and longevity. This requires a delicate balance of exogenous insulin administration, meticulous glucose monitoring, nutritional planning, regular physical activity, and robust psychosocial support.

5.1. Insulin Therapy

Insulin therapy is the absolute cornerstone of T1D management, as the body can no longer produce its own. The aim is to mimic the physiological pattern of insulin secretion from a healthy pancreas, which involves both a continuous basal insulin supply and bolus doses for meals and correction of high glucose levels [40].

  • Types of Insulin: Insulins are categorized by their onset, peak, and duration of action:

    • Rapid-acting insulin analogs (e.g., aspart, lispro, glulisine): Onset within 5-15 minutes, peak at 30-90 minutes, duration 3-5 hours. Used for mealtime boluses and correction of hyperglycemia.
    • Short-acting insulin (Regular insulin): Onset 30-60 minutes, peak 2-4 hours, duration 5-8 hours. Less commonly used for boluses due to slower action.
    • Intermediate-acting insulin (NPH): Onset 2-4 hours, peak 4-10 hours, duration 10-16 hours. Used for basal coverage, but often replaced by long-acting analogs.
    • Long-acting insulin analogs (e.g., glargine, detemir, degludec): Onset 1-2 hours, minimal to no peak, duration 18-42 hours. Provide continuous basal insulin coverage.
    • Ultra-long-acting insulin (e.g., glargine U300, degludec U200): Provide even longer and flatter profiles, suitable for 24-hour basal coverage.
  • Insulin Delivery Methods:

    • Multiple Daily Injections (MDI): This involves using long-acting insulin (once or twice daily) for basal coverage and rapid-acting insulin (before meals and for corrections) via insulin pens or syringes. It offers flexibility but requires multiple injections daily [41].
    • Continuous Subcutaneous Insulin Infusion (CSII) / Insulin Pumps: An insulin pump is a small, computerized device worn externally that delivers rapid-acting insulin continuously through a catheter inserted under the skin. It provides a highly customizable basal rate and allows for precise bolus dosing. Modern pumps offer features like temporary basal rates, extended boluses, and integration with continuous glucose monitors (CGM) [42].
    • Hybrid Closed-Loop Systems (Automated Insulin Delivery – AID): These systems, often termed ‘artificial pancreas’ systems, represent a significant advancement. They integrate an insulin pump with a CGM and an algorithm that automatically adjusts basal insulin delivery in response to real-time glucose readings, with user input required for meal boluses. Examples include the MiniMed™ 670G/770G/780G, t:slim X2™ with Control-IQ™, and Omnipod 5®. These systems have been shown to improve glycemic control (e.g., increased time in range) and reduce the burden of diabetes management [43].
    • Future of AID: Fully automated closed-loop systems (requiring no user input for meals) and bi-hormonal systems (delivering both insulin and glucagon) are under development.

5.2. Glucose Monitoring

Accurate and frequent glucose monitoring is fundamental to safe and effective T1D management.

  • Self-Monitoring of Blood Glucose (SMBG): This involves pricking a finger to obtain a blood sample for analysis with a glucose meter. It provides instantaneous glucose values at specific time points, informing insulin doses and identifying hypo/hyperglycemia. However, it only provides snapshots [44].
  • Continuous Glucose Monitoring (CGM): CGM systems utilize a small sensor inserted under the skin (typically on the arm or abdomen) to measure interstitial glucose levels every few minutes, providing real-time data or ‘flash’ data (scanned periodically). CGMs offer numerous advantages:
    • Trend Information: Displaying glucose trends (rising, falling, stable) helps predict future glucose excursions.
    • Reduced Hypo/Hyperglycemia: Alerts for high or low glucose levels enable timely intervention.
    • Time in Range (TIR): A key metric indicating the percentage of time glucose is within the target range (e.g., 70-180 mg/dL), increasingly recognized as a better indicator of glycemic control than HbA1c alone [45].
    • Data Analysis: Downloadable data allows healthcare providers and patients to identify patterns, optimize insulin regimens, and improve decision-making.

5.3. Nutritional Management and Carbohydrate Counting

Dietary management is paramount for individuals with T1D to align insulin delivery with carbohydrate intake. Carbohydrate counting is a cornerstone skill, allowing patients to estimate the carbohydrate content of meals and snacks and adjust their rapid-acting insulin doses accordingly. This typically involves using an ‘insulin-to-carbohydrate ratio’ [46].

  • Meal Planning: Focus is on balanced meals with appropriate proportions of carbohydrates, proteins, and fats.
  • Glycemic Index (GI)/Glycemic Load (GL): Understanding how different carbohydrates affect blood glucose (rapid vs. slow rise) can aid in meal planning, though strict adherence to low-GI diets is not always necessary if insulin dosing is adjusted appropriately.
  • Fiber Intake: Encouraging adequate fiber intake helps with satiety and can modulate glucose absorption.
  • Fat and Protein Considerations: While insulin doses are primarily based on carbohydrates, high-fat and high-protein meals can also influence glucose levels, sometimes requiring adjustments to insulin timing or dosage [47].

5.4. Physical Activity

Regular physical activity is highly beneficial for overall health, cardiovascular fitness, and insulin sensitivity in individuals with T1D. However, exercise management requires careful planning due to its profound effect on blood glucose levels. Exercise can increase glucose uptake by muscles and improve insulin sensitivity, leading to a risk of hypoglycemia, particularly during and several hours after activity [48].

  • Pre-exercise Adjustments: Reducing insulin doses (basal and/or bolus) and/or consuming additional carbohydrates before, during, or after exercise.
  • Monitoring: Frequent glucose monitoring (especially with CGM) is crucial during and after exercise to identify trends and prevent hypo- or hyperglycemia.
  • Hydration: Adequate hydration is important, especially during prolonged or intense exercise.
  • Carbohydrate Availability: Having fast-acting carbohydrates readily available to treat hypoglycemia.

5.5. Education and Psychosocial Support

Comprehensive diabetes education is indispensable for empowering individuals with T1D and their families to self-manage the condition effectively. This includes understanding insulin types, injection techniques, pump operation, carbohydrate counting, sick day rules, and complication prevention [49].

  • Diabetes Distress and Burnout: Living with a chronic condition like T1D can lead to significant psychological burden, including diabetes distress, anxiety, depression, and ‘burnout,’ where individuals feel overwhelmed and disengage from self-management [50].
  • Psychological Support: Access to mental health professionals specializing in chronic illness is crucial to address these challenges, promote coping strategies, and enhance adherence to management plans.
  • Peer Support: Connecting with others living with T1D can provide invaluable emotional support, shared experiences, and practical advice.
  • Family Involvement: For children and adolescents, active family involvement in education and management is critical for successful outcomes.

5.6. Technological Advancements and Future Directions

The field of T1D management is rapidly evolving with technological innovations:

  • Smart Insulin Pens: These devices record insulin doses, time, and integrate with apps to provide insights and assist with calculations.
  • Integrated Digital Health Platforms: Apps and software that integrate data from CGMs, pumps, and activity trackers to provide a holistic view of diabetes management, facilitating data sharing with healthcare providers.
  • Non-invasive Glucose Monitoring: Research continues into non-invasive methods, though none are currently clinically validated for accuracy compared to blood-based or interstitial fluid measurements.
  • Encapsulated Beta Cells: A long-term goal for beta-cell replacement, where engineered or donated beta cells are enclosed in a protective membrane, allowing insulin secretion while preventing immune rejection [51].

Many thanks to our sponsor Esdebe who helped us prepare this research report.

6. Complications of Type 1 Diabetes

Chronic exposure to hyperglycemia, if uncontrolled, can lead to a wide array of severe and potentially debilitating acute and long-term complications, affecting virtually every organ system. These complications underscore the critical importance of stringent glycemic control.

6.1. Acute Complications

Acute complications are typically rapid in onset and require immediate medical intervention.

  • Diabetic Ketoacidosis (DKA):

    • Pathophysiology: DKA results from absolute insulin deficiency, leading to unchecked lipolysis and increased counter-regulatory hormones (glucagon, catecholamines, cortisol, growth hormone). This stimulates gluconeogenesis and glycogenolysis, causing severe hyperglycemia. The breakdown of fats produces ketone bodies (beta-hydroxybutyrate, acetoacetate, acetone), which are acidic, leading to metabolic acidosis. Severe dehydration ensues from osmotic diuresis [39].
    • Clinical Presentation: Polyuria, polydipsia, weight loss, nausea, vomiting, abdominal pain, Kussmaul respirations (deep, labored breathing), fruity breath (due to acetone), and altered mental status (from lethargy to coma).
    • Diagnosis: Hyperglycemia (blood glucose >250 mg/dL), ketonemia/ketonuria, and metabolic acidosis (pH <7.3, bicarbonate <18 mEq/L).
    • Management: Urgent intravenous fluid resuscitation, insulin administration (typically IV infusion), electrolyte replacement (especially potassium), and close monitoring in an intensive care setting.
  • Severe Hypoglycemia (Insulin Shock):

    • Causes: Excess insulin, insufficient carbohydrate intake, unexpected or intense physical activity, delayed meals, or alcohol consumption.
    • Symptoms:
      • Autonomic (adrenergic): Shakiness, sweating, palpitations, anxiety, hunger, nausea, tingling.
      • Neuroglycopenic: Headache, confusion, irritability, blurred vision, dizziness, difficulty speaking, weakness, loss of consciousness, seizures, coma [52].
    • Classification:
      • Level 1 (Glucose Alert Value): Glucose <70 mg/dL (3.9 mmol/L).
      • Level 2 (Clinically Significant Hypoglycemia): Glucose <54 mg/dL (3.0 mmol/L).
      • Level 3 (Severe Hypoglycemia): Requires assistance from another person for recovery due to altered mental and/or physical status.
    • Management: Prompt ingestion of fast-acting carbohydrates (e.g., glucose tablets, juice), followed by a complex carbohydrate. For severe hypoglycemia, glucagon injection (intramuscular or nasal spray) is necessary to raise blood glucose quickly.
  • Hyperosmolar Hyperglycemic State (HHS): While far more common in T2D, HHS can rarely occur in T1D, particularly in older individuals with some residual beta-cell function or in those who are severely ill. It is characterized by extreme hyperglycemia (>600 mg/dL), severe dehydration, and hyperosmolarity, without significant ketosis. Management involves aggressive fluid resuscitation, insulin, and electrolyte correction [53].

6.2. Chronic Complications

Chronic complications develop over years of sustained hyperglycemia, leading to damage to blood vessels and nerves throughout the body. They are broadly categorized into microvascular (small vessel) and macrovascular (large vessel) complications.

  • Microvascular Complications: Result from damage to small blood vessels.

    • Diabetic Retinopathy: Damage to the blood vessels in the retina, the light-sensitive tissue at the back of the eye. It is the leading cause of blindness in working-age adults.
      • Stages: Non-proliferative (early stage, microaneurysms, hemorrhages, exudates) and Proliferative (advanced stage, new, fragile blood vessels grow on the retina, leading to vitreous hemorrhage and retinal detachment) [54]. Macular edema can occur at any stage.
      • Screening: Annual dilated eye exams are crucial.
      • Treatment: Strict glycemic and blood pressure control. Laser photocoagulation, anti-vascular endothelial growth factor (anti-VEGF) injections, and vitrectomy are treatments for advanced retinopathy.
    • Diabetic Nephropathy: Damage to the kidneys’ filtering units (glomeruli), leading to impaired kidney function and eventually end-stage renal disease (ESRD), requiring dialysis or kidney transplantation.
      • Stages: Characterized by progressive albuminuria (protein in urine) and decline in glomerular filtration rate (GFR).
      • Screening: Annual screening for albuminuria (urine albumin-to-creatinine ratio, ACR) and monitoring of estimated GFR (eGFR) [55].
      • Management: Optimal glycemic control, blood pressure control (ACE inhibitors or ARBs are first-line), and increasingly, SGLT2 inhibitors and GLP-1 receptor agonists have shown kidney protective benefits [56].
    • Diabetic Neuropathy: Damage to nerves throughout the body, leading to a variety of symptoms depending on the affected nerves.
      • Peripheral Neuropathy: Most common, affecting nerves in the legs and feet, causing numbness, tingling, burning pain, and loss of sensation. This increases the risk of foot ulcers and amputations due to unrecognized injuries [57].
      • Autonomic Neuropathy: Affects nerves controlling internal organs, leading to diverse symptoms:
        • Cardiovascular: Orthostatic hypotension, silent myocardial ischemia.
        • Gastrointestinal: Gastroparesis (delayed stomach emptying leading to nausea, vomiting, early satiety), diarrhea, constipation.
        • Genitourinary: Erectile dysfunction, bladder dysfunction.
      • Diagnosis: Clinical symptoms, neurological examination, nerve conduction studies.
      • Management: Strict glycemic control is the only way to prevent progression. Symptomatic management (e.g., pain relief with gabapentinoids, tricyclic antidepressants for neuropathic pain; prokinetics for gastroparesis).
  • Macrovascular Complications: Affect large blood vessels, leading to accelerated atherosclerosis and cardiovascular diseases, which are the leading cause of morbidity and mortality in T1D [58].

    • Coronary Artery Disease (CAD): Increased risk of heart attacks and angina.
    • Stroke: Increased risk of ischemic stroke.
    • Peripheral Artery Disease (PAD): Reduced blood flow to the limbs, particularly the legs and feet, contributing to non-healing ulcers and amputations.
    • Pathophysiology: Chronic hyperglycemia, dyslipidemia, hypertension, and systemic inflammation contribute to endothelial dysfunction, plaque formation, and vascular stiffness. T1D patients often have a higher burden of cardiovascular risk factors and may experience cardiovascular events at an earlier age and with less typical symptoms (e.g., ‘silent’ heart attacks due to autonomic neuropathy) [59].
    • Management: Aggressive management of traditional cardiovascular risk factors: glycemic control, blood pressure control, lipid management (statins), antiplatelet therapy (aspirin if indicated), smoking cessation, and lifestyle modifications.
  • Other Complications and Comorbidities: T1D patients are at higher risk for other autoimmune diseases due to shared genetic predispositions:

    • Autoimmune Thyroid Disease: Hypothyroidism (Hashimoto’s thyroiditis) or hyperthyroidism (Graves’ disease) is common, warranting regular screening for thyroid function [60].
    • Celiac Disease: An autoimmune disorder triggered by gluten, affecting the small intestine. Screening is recommended due to higher prevalence in T1D patients [61].
    • Addison’s Disease: Rare but serious autoimmune adrenal insufficiency.
    • Osteoporosis: Increased risk of reduced bone mineral density and fractures, possibly due to chronic inflammation and insulin deficiency.
    • Depression and Anxiety: Higher rates of mental health disorders, often linked to the chronic burden of disease management [50].
    • Infections: Impaired immune function and high glucose levels increase susceptibility to various infections (e.g., skin, urinary tract, respiratory), including severe infections like mucormycosis.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

7. Ongoing Research into Prevention and Potential Cures

The profound impact of T1D and its complications drives extensive global research aimed at preventing the disease and ultimately finding a cure. These efforts span multiple domains, from understanding initial immune responses to replacing lost beta cells.

7.1. Immunomodulatory Therapies

Immunomodulatory strategies aim to halt or reverse the autoimmune destruction of beta cells, particularly in the early stages of the disease (pre-symptomatic or newly diagnosed). The goal is to re-establish immune tolerance without compromising the body’s overall ability to fight infections.

  • Anti-CD3 Monoclonal Antibodies (e.g., Teplizumab, Otelixizumab): These antibodies target the CD3 receptor on T cells, inducing transient T-cell anergy or deletion and modifying T-cell responses. Teplizumab was the first drug approved in the United States to delay the onset of T1D in individuals aged 8 years and older with Stage 2 T1D, by an average of about two years [62]. This landmark approval validated the concept of immune modulation in T1D prevention.
  • Abatacept (CTLA4-Ig): A fusion protein that blocks co-stimulation required for full T-cell activation. It has shown some promise in preserving beta-cell function in new-onset T1D [63].
  • Anti-CD20 Monoclonal Antibodies (e.g., Rituximab): Target CD20 on B cells, leading to B-cell depletion. B cells are important in T1D as antigen-presenting cells and for autoantibody production. Rituximab has shown some initial benefits in preserving beta-cell function [64].
  • JAK Inhibitors (e.g., Tofacitinib): These small molecules inhibit Janus kinases, which are involved in cytokine signaling pathways that drive inflammation. They are being investigated for their potential to modulate the immune response in T1D [65].
  • Cytokine Modulation: Therapies targeting specific pro-inflammatory cytokines (e.g., IL-1, IL-6, TNF-α) or enhancing regulatory cytokines (e.g., IL-10) are under investigation to shift the immune balance away from destructive inflammation [66].
  • Antigen-Specific Immunotherapy: Aims to re-educate the immune system to tolerate specific beta-cell autoantigens (e.g., proinsulin, GAD65) by administering the antigen in a non-immunogenic way, often through peptides or modified cells, to induce tolerance [67].

Challenges for immunomodulatory therapies include identifying the optimal timing of intervention (early stages are best), maintaining long-term efficacy, and avoiding broad immunosuppression.

7.2. Beta-Cell Replacement Strategies

For individuals with established T1D, the goal is to replace the destroyed beta cells and restore endogenous insulin production, thereby eliminating the need for exogenous insulin and improving glycemic control.

  • Islet Transplantation: Involves transplanting insulin-producing islet cells, typically from cadaveric human pancreases, into the portal vein of the recipient. This procedure can restore insulin production, but it requires lifelong immunosuppression to prevent rejection, which carries significant side effects. It is currently limited by the scarcity of donor organs and the need for chronic immunosuppression, making it a viable option for a select few with severe glycemic lability or renal failure [68].
  • Stem Cell Therapy: This area holds immense promise.

    • Embryonic Stem Cells (ESCs) and Induced Pluripotent Stem Cells (iPSCs): These cells can be differentiated into functional insulin-producing beta cells in vitro. Clinical trials are underway to test the safety and efficacy of implanting these lab-grown beta cells [69].
    • Encapsulation Strategies: To overcome the need for systemic immunosuppression, researchers are developing micro- or macro-encapsulation devices that protect transplanted islets or stem-cell-derived beta cells from immune attack while allowing glucose and insulin exchange. This creates an ‘immunoisolation’ barrier [51].
    • Gene Editing for Immune Evasion: Advances in CRISPR/Cas9 technology allow for genetic modification of stem cell-derived beta cells to make them ‘invisible’ to the immune system, potentially eliminating the need for immunosuppression [70].
  • Beta-Cell Regeneration: Research is exploring approaches to stimulate the regeneration of existing beta cells within the pancreas or to induce differentiation of other pancreatic cell types into insulin-producing cells. This includes targeting pathways involved in beta-cell proliferation or transdifferentiation [71].

7.3. Vaccines

Vaccine research in T1D is diverse:

  • Antigen-Specific Vaccines: Similar to antigen-specific immunotherapy, these aim to induce immune tolerance to specific beta-cell autoantigens, essentially ‘tolerizing’ the immune system to self-antigens [67].
  • Viral Vaccines: Developing vaccines against implicated environmental triggers, such as enteroviruses (e.g., Coxsackievirus B), could potentially prevent the initial autoimmune trigger in susceptible individuals [13].

7.4. Gene Therapy

Gene therapy involves introducing, altering, or removing genetic material within cells to treat a disease. In T1D, this could involve:

  • Modifying Immune Cells: Engineering immune cells (e.g., T cells) to become tolerogenic or to express factors that protect beta cells.
  • Expressing Insulin in Non-Beta Cells: Programming other cell types in the body (e.g., hepatocytes, muscle cells) to produce insulin in a glucose-responsive manner, effectively creating alternative insulin factories [72].

7.5. Biomarker Discovery and Prediction

Ongoing efforts focus on identifying novel biomarkers beyond traditional autoantibodies that can more accurately predict T1D onset, monitor disease progression, and identify individuals most likely to respond to specific preventive therapies. This includes advanced autoantibody panels, genetic risk scores, metabolic markers (e.g., proinsulin-to-C-peptide ratio), and immune cell phenotyping [34, 73].

Many thanks to our sponsor Esdebe who helped us prepare this research report.

8. Impact of COVID-19 on New-Onset Type 1 Diabetes and Existing T1D Management

The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has created a complex landscape for individuals with diabetes, particularly those with T1D. Emerging evidence has suggested a potential association between SARS-CoV-2 infection and the incidence of new-onset T1D, as well as significant challenges in managing existing T1D [74].

8.1. Increase in New-Onset Cases

Several observational studies and registries from various countries have reported an increase in the incidence of new-onset T1D, particularly in children and adolescents, during the COVID-19 pandemic compared to pre-pandemic periods.

  • A multi-center analysis in the United States, for instance, found a significant rise in newly diagnosed T1D patients in 2020 compared to the previous year, with a notable increase in patients presenting in diabetic ketoacidosis (DKA) during the pandemic [1].
  • Similar trends have been observed in European countries, including Germany, the UK, and Italy, reporting a higher incidence of T1D in pediatric populations [75, 76]. Some studies have also pointed to a more severe presentation at diagnosis, with higher rates of DKA, potentially attributable to delayed diagnosis and fear of seeking medical care during lockdowns [77].

While an increase in new-onset cases has been reported, it is crucial to interpret these findings cautiously. Not all studies have shown a definitive, universal increase, and some conflicting data exist [78]. The observed increase might be influenced by multiple factors, including:

  • Direct Viral Effects: SARS-CoV-2 utilizes the Angiotensin-converting enzyme 2 (ACE2) receptor for cell entry, which is expressed on pancreatic beta cells [79]. This raises the hypothesis of direct viral tropism and cytopathic effects on beta cells, potentially leading to inflammation (insulitis) and dysfunction, or even destruction, similar to other viruses implicated in T1D pathogenesis.
  • Indirect Immune Dysregulation: The profound systemic inflammation and ‘cytokine storm’ associated with severe COVID-19 can exacerbate existing autoimmune predispositions or trigger new ones. Pro-inflammatory cytokines (e.g., IL-6, TNF-alpha) are known to be detrimental to beta-cell function and survival [80]. The infection could also lead to molecular mimicry, where viral antigens resemble beta-cell antigens, prompting an autoimmune response.
  • Environmental and Lifestyle Changes: Pandemic-related lockdowns and changes in lifestyle (e.g., reduced physical activity, altered dietary habits, increased stress, altered microbial exposure) might have contributed to an accelerated onset of T1D in genetically predisposed individuals. Reduced exposure to common childhood infections (due to lockdowns) might also influence immune system development in a way that aligns with the hygiene hypothesis [81].
  • Delayed Diagnosis: Healthcare disruptions and public reluctance to seek medical attention during the pandemic likely led to delays in diagnosis for some children, resulting in more advanced symptoms and higher rates of DKA at presentation. This could inflate the reported incidence of ‘severe’ new-onset cases [77].

It is important to note that direct, conclusive evidence that SARS-CoV-2 directly causes T1D remains elusive. While several case reports describe new-onset T1D following COVID-19, establishing a definitive causal link requires further robust epidemiological and mechanistic studies. Many experts consider it more likely that the virus acts as a ‘trigger’ in genetically susceptible individuals, accelerating or unmasking pre-existing autoimmunity, rather than initiating the disease de novo in previously unaffected individuals [82].

8.2. Management of Existing T1D Patients During COVID-19

The pandemic presented substantial challenges for individuals already living with T1D, impacting their glycemic control, access to care, and mental well-being.

  • Increased Risk of Severe Outcomes: Individuals with diabetes, including T1D, were identified as being at higher risk for severe COVID-19 outcomes, including hospitalization, intensive care unit admission, and death [83]. This was largely attributed to the pro-inflammatory and pro-thrombotic state associated with uncontrolled diabetes, which exacerbates the systemic inflammatory response to SARS-CoV-2.
  • Sick Day Management: Managing T1D during any acute illness requires careful monitoring of blood glucose and ketones, with potential increases in insulin requirements. COVID-19, with its varied symptoms and potential for severe inflammation, added complexity to ‘sick day’ management, necessitating close communication with healthcare teams [84].
  • Access to Care and Supplies: Lockdowns and healthcare system strain led to disruptions in routine appointments, reduced access to diabetes educators and dietitians, and potential difficulties in obtaining insulin and other supplies. Telemedicine rapidly expanded to fill this gap, proving essential for continuity of care [85].
  • Psychological Burden: The fear of infection, social isolation, economic uncertainties, and the added pressure of managing a chronic condition during a pandemic significantly increased anxiety, stress, and diabetes distress among T1D patients and their families [86].
  • Vaccine Considerations: T1D patients were prioritized for COVID-19 vaccination due to their increased risk, and evidence has largely shown the vaccines to be safe and effective in this population, without significant adverse effects on glycemic control [87].

Many thanks to our sponsor Esdebe who helped us prepare this research report.

9. Conclusion

Type 1 diabetes remains a complex and formidable global health challenge, profoundly impacting the lives of millions worldwide. Its intricate pathogenesis, arising from a delicate interplay of genetic predispositions, environmental triggers, and immune system dysregulation, continues to be a focal point of intense scientific inquiry. While there has been remarkable progress in understanding the disease mechanisms, the relentless autoimmune destruction of beta cells still necessitates lifelong, meticulous management involving exogenous insulin administration, advanced glucose monitoring technologies, and comprehensive lifestyle adaptations.

Contemporary management paradigms, bolstered by innovations such as advanced automated insulin delivery systems, continuous glucose monitoring, and sophisticated nutritional counseling, have significantly improved glycemic control, reduced the burden of management, and consequently mitigated the risk of both acute and chronic complications. Nevertheless, the specter of severe hypoglycemia, diabetic ketoacidosis, and long-term microvascular and macrovascular complications underscores the critical need for continued vigilance and ongoing patient education.

The global health crisis presented by the COVID-19 pandemic has introduced new dimensions to the T1D landscape. While the precise causal link between SARS-CoV-2 infection and new-onset T1D remains a subject of ongoing investigation and debate, the observed increase in incident cases and the heightened severity of presentation highlight the potential for viral infections to serve as environmental triggers or accelerators in genetically susceptible individuals. Furthermore, the pandemic profoundly impacted the delivery and experience of care for individuals living with established T1D, necessitating rapid adaptations in healthcare delivery models and bringing to light the substantial psychological burden associated with managing a chronic condition amidst a global crisis.

Looking ahead, the horizon of T1D research is bright with promise. Groundbreaking advancements in immunomodulatory therapies, such as the approval of teplizumab, offer tangible hope for delaying or even preventing disease onset in at-risk individuals. Concurrently, rapid progress in beta-cell replacement strategies, including advanced islet transplantation and the burgeoning field of stem cell-derived beta cell therapy, coupled with innovative encapsulation techniques and gene editing approaches, holds the potential to offer curative solutions for those already living with T1D. Continued research into biomarker discovery, antigen-specific immunotherapies, and viral vaccines represents concerted efforts to address the disease at its root.

In conclusion, T1D is a dynamic field where scientific understanding and clinical management are continuously evolving. The collective efforts of researchers, clinicians, and patients, informed by lessons from global health events like the COVID-19 pandemic, are paving the way for more effective prevention strategies, improved therapeutic outcomes, and, ultimately, the aspiration of a world free from the burden of type 1 diabetes.

Many thanks to our sponsor Esdebe who helped us prepare this research report.

References

  1. Wolf, S. E., et al. (2022). Increase in newly diagnosed type 1 diabetes in youth during the COVID‐19 pandemic in the United States: A multi‐center analysis. Pediatric Diabetes, 23(1), 38-46. onlinelibrary.wiley.com
  2. Patterson, C. C., et al. (2019). The worldwide incidence of type 1 diabetes in the 21st century: a systematic review and meta-analysis. Diabetes Care, 42(6), 1035-1044.
  3. Atkinson, M. A., & Eisenbarth, G. S. (2001). Type 1 diabetes: new perspectives on disease pathogenesis and treatment. The Lancet, 358(9277), 221-229.
  4. Insel, R. A., et al. (2015). Staging preclinical type 1 diabetes: a Scientific Statement of the American Diabetes Association, JDRF, and the Endocrine Society. Diabetes Care, 38(7), 1361-1374.
  5. Pociot, F., & Lernmark, Å. (2016). Genetic risk factors for type 1 diabetes. Diabetes Care, 39(Suppl 2), S160-S169.
  6. Erlich, H., et al. (2008). HLA DR-DQ haplotypes and alleles as markers of type 1 diabetes risk: a prospective study of 2,238 Caucasian newborns. Diabetes, 57(4), 1085-1092.
  7. Bottini, N., et al. (2004). A functional variant of PTPN22 is associated with an increased risk of type 1 diabetes in a Caucasian population. Nature Genetics, 36(11), 1195-1200.
  8. Vafiadis, P., et al. (1997). Insulin expression in human thymus is modulated by INS VNTR genotypes. Nature Genetics, 15(3), 289-292.
  9. Nistico, L., et al. (1996). The CTLA-4 gene region of chromosome 2q33 is linked to, and associated with, type 1 diabetes. Human Molecular Genetics, 5(7), 1075-1080.
  10. Smyth, D. J., et al. (2006). The interferon-induced helicase IFIH1 is a major gene for type 1 diabetes. Nature Genetics, 38(6), 617-619.
  11. Wallace, C., et al. (2010). Genome-wide association study identifies genes for the PI3K pathway and immune activation in type 1 diabetes. Nature Genetics, 42(9), 767-772.
  12. Wilkin, T. J. (2001). The accelerator hypothesis: a review of the evidence for the unity of Type 1 and Type 2 diabetes. Diabetologia, 44(8), 917-925.
  13. Rewers, M., & Ludvigsson, J. (2016). Environmental risk factors for type 1 diabetes. Lancet, 387(10035), 2340-2348.
  14. Filippi, C. M., & von Herrath, M. G. (2008). Viral trigger for type 1 diabetes: pros and cons. Diabetes Care, 31(Suppl 2), S172-S178.
  15. Vaarala, O., et al. (2000). Cow’s milk beta-casein and type 1 diabetes. Annals of Medicine, 32(8), 536-542.
  16. Lundin, K. E. A., & Sollid, L. M. (2015). Celiac disease and type 1 diabetes: towards an understanding of the molecular basis of their association. Current Opinion in Gastroenterology, 31(6), 464-471.
  17. Hyppönen, E., et al. (2008). Intake of vitamin D and risk of type 1 diabetes: a birth-cohort study. The Lancet, 360(9337), 1309-1311.
  18. Ludvigsson, J., et al. (2020). The gut microbiome and type 1 diabetes. Nature Reviews Endocrinology, 16(11), 606-620.
  19. Bluestone, J. A., et al. (2010). Type 1 diabetes: as sweet as it gets. Nature Medicine, 16(11), 1279-1282.
  20. Maahs, D. M., et al. (2010). Type 1 diabetes. The Lancet, 376(9742), 1153-1165.
  21. van Belle, T. L., et al. (2011). The pancreatic beta cell in type 1 diabetes: a target for a multifaceted attack. Physiological Reviews, 91(1), 133-171.
  22. Eisenbarth, G. S. (1986). Type I diabetes mellitus. A chronic autoimmune disease. The New England Journal of Medicine, 314(21), 1360-1368.
  23. Brusko, T. M., et al. (2008). The role of FOXP3+ regulatory T cells in type 1 diabetes. Endocrine Reviews, 29(4), 438-456.
  24. Pugliese, A. (2017). Insulitis in type 1 diabetes: still an enigma. Diabetes, 66(1), 1-13.
  25. Eizirik, D. L., et al. (2013). Endoplasmic reticulum stress and the unfolded protein response in pancreatic beta-cells: the good, the bad, and the ugly. Endocrine Reviews, 34(3), 462-491.
  26. Redondo, M. J., et al. (2018). The genetics of type 1 diabetes: a guide for the clinician. The Journal of Clinical Endocrinology & Metabolism, 103(12), 4381-4399.
  27. Kyvik, K. O., et al. (1995). Concordance rates of Type 1 diabetes in Danish twins: from a national Twin Register. BMJ, 311(7010), 913-917.
  28. Pociot, F., & Lernmark, Å. (2016). Genetic risk factors for type 1 diabetes. Diabetes Care, 39(Suppl 2), S160-S169.
  29. DIAMOND Project Group. (2006). Incidence and trends of childhood Type 1 diabetes worldwide 1990-1999. Diabetic Medicine, 23(8), 857-866.
  30. Patterson, C. C., et al. (2019). The worldwide incidence of type 1 diabetes in the 21st century: a systematic review and meta-analysis. Diabetes Care, 42(6), 1035-1044.
  31. Lipton, R. B., et al. (1995). The epidemiology of IDDM in the U.S. Diabetes Care, 18(11), 1510-1521.
  32. Maahs, D. M., et al. (2010). Type 1 diabetes. The Lancet, 376(9742), 1153-1165.
  33. Bach, J. F. (2002). The effect of infections on susceptibility to autoimmune and allergic diseases. The New England Journal of Medicine, 347(13), 911-921.
  34. Krischer, J. P., et al. (2015). The environmental determinants of diabetes in the young (TEDDY) study. Current Opinion in Endocrinology, Diabetes and Obesity, 22(4), 282-288.
  35. Daneman, D. (2006). Type 1 diabetes. The Lancet, 367(9513), 847-858.
  36. American Diabetes Association. (2024). Standards of Care in Diabetes—2024. Diabetes Care, 47(Suppl 1), S1-S302.
  37. Sosenko, J. M., et al. (2012). The natural history of the development of type 1 diabetes in the Diabetes Prevention Trial-Type 1. Diabetes Care, 35(3), 596-602.
  38. Hattersley, A. T., & Pearson, E. R. (2006). Monogenic diabetes in the 21st century. The Lancet, 367(9516), 1317-1327.
  39. Kitabchi, A. E., et al. (2009). Hyperglycemic crises in adult patients with diabetes. Diabetes Care, 32(7), 1335-1343.
  40. American Diabetes Association. (2024). Standards of Care in Diabetes—2024. Diabetes Care, 47(Suppl 1), S1-S302.
  41. Hirsch, I. B. (2005). Insulin analogs. The New England Journal of Medicine, 352(2), 174-184.
  42. Bode, B. W., & Tamborlane, W. V. (2002). Insulin pumps: advances in technology and utilization. Endocrinology and Metabolism Clinics of North America, 31(3), 577-589.
  43. Bergenstal, R. M., et al. (2021). The Advanced Hybrid Closed-Loop System in Adolescents and Adults with Type 1 Diabetes. Diabetes Technology & Therapeutics, 23(Suppl 1), S12-S19.
  44. American Diabetes Association. (2024). Standards of Care in Diabetes—2024. Diabetes Care, 47(Suppl 1), S1-S302.
  45. Battelino, T., et al. (2019). Clinical Targets for Continuous Glucose Monitoring-Data Interpretation: Recommendations From the International Consensus on Time in Range. Diabetes Care, 42(8), 1593-1603.
  46. Warshaw, H. S., & Powers, M. A. (2019). The 2018 American Diabetes Association Nutrition Recommendations: Guide for Registered Dietitian Nutritionists. Journal of the Academy of Nutrition and Dietetics, 119(9), 1622-1629.
  47. Bao, J., et al. (2011). Food insulin index: a new concept in diabetes dietetics. Journal of Functional Foods, 3(3), 143-154.
  48. Riddell, M. C., et al. (2017). Exercise and type 1 diabetes: a consensus statement from the American Diabetes Association. Diabetes Care, 40(11), 1590-1600.
  49. Powers, M. A., et al. (2017). Diabetes self-management education and support in type 2 diabetes: A joint position statement of the American Diabetes Association, the American Association of Diabetes Educators, and the Academy of Nutrition and Dietetics. Diabetes Care, 40(Suppl 1), S104-S113.
  50. Polonsky, W. H., et al. (1995). Diabetes distress in type 1 diabetes: prevalence, clinical correlates, and predictors. Diabetes Care, 18(1), 101-107.
  51. Shapiro, A. M., et al. (2017). Encapsulated cell therapies for diabetes. Science Translational Medicine, 9(378), eaah4839.
  52. International Hypoglycaemia Study Group. (2017). Glucose monitoring, clinical targets, and hypoglycaemia in type 1 diabetes: a review. Diabetologia, 60(8), 1530-1538.
  53. Gosmanov, A. R., & Kitabchi, A. E. (2013). Diabetic ketoacidosis and hyperosmolar hyperglycemic state. Endocrinology and Metabolism Clinics of North America, 42(4), 721-739.
  54. Frank, R. N. (2004). Diabetic retinopathy. The New England Journal of Medicine, 350(1), 48-58.
  55. Alicic, R. Z., et al. (2017). Diabetic kidney disease: challenges, progress, and possibilities. Clinical Journal of the American Society of Nephrology, 12(12), 2032-2045.
  56. Wanner, C., et al. (2016). Empagliflozin and Progression of Kidney Disease in Type 2 Diabetes. The New England Journal of Medicine, 375(4), 323-334.
  57. Boulton, A. J., et al. (2005). Diabetic neuropathies: a statement by the American Diabetes Association. Diabetes Care, 28(4), 956-962.
  58. Haffner, S. M., et al. (1998). Mortality from coronary heart disease in subjects with type 1 and type 2 diabetes: a population-based study. Diabetes Care, 21(9), 1278-1282.
  59. Piconi, S., et al. (2003). Oxidative stress in diabetes. Journal of Biological Chemistry, 278(41), 40049-40058.
  60. Kordonouri, O., et al. (2004). Autoimmune thyroiditis in children and adolescents with type 1 diabetes mellitus. Journal of Pediatric Endocrinology and Metabolism, 17(Suppl 1), 603-608.
  61. Mohn, A., et al. (2005). Celiac disease in children with type 1 diabetes mellitus: a multicenter study. Pediatrics, 115(3), e345-e349.
  62. Herold, K. C., et al. (2019). An anti-CD3 antibody, teplizumab, in relatives at risk for type 1 diabetes. The New England Journal of Medicine, 381(7), 603-613.
  63. Orban, T., et al. (2014). Costimulation modulation with abatacept in patients with recent-onset type 1 diabetes: a multicentre, randomised, double-blind, placebo-controlled trial. The Lancet, 383(9928), 1629-1638.
  64. Pescovitz, M. D., et al. (2009). Rituximab in recent-onset type 1 diabetes mellitus. The New England Journal of Medicine, 360(11), 1083-1097.
  65. O’Shea, J. J., & Plenge, W. H. (2012). JAK and STAT signaling molecules in inflammation and autoimmunity. Nature Reviews Drug Discovery, 11(10), 743-756.
  66. Eizirik, D. L., & Mandrup-Poulsen, T. (2001). A role for the interleukin-1β system in the pathogenesis of type 1 diabetes. Diabetologia, 44(Suppl 3), B106-B114.
  67. Peakman, M., et al. (2011). Peptide vaccination in type 1 diabetes. Expert Review of Vaccines, 10(7), 963-971.
  68. Shapiro, A. M., et al. (2000). Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. The New England Journal of Medicine, 343(4), 230-238.
  69. Pagliuca, F. W., et al. (2014). Generation of functional human pancreatic beta cells in vitro. Cell, 159(2), 428-439.
  70. De Bortoli, M., et al. (2020). Engineering Hypoimmune Allogeneic Stem Cell Therapies. Trends in Biotechnology, 38(4), 438-450.
  71. Melton, D. A. (2012). Stem cells and diabetes: challenges and opportunities. Cell Stem Cell, 11(6), 724-728.
  72. Gontarczyk, M., et al. (2021). Current and future prospects for gene therapy in diabetes. Current Opinion in Endocrinology, Diabetes and Obesity, 28(5), 478-485.
  73. Lernmark, Å., et al. (2021). The TEDDY study: a decade of insights into type 1 diabetes risk. Nature Reviews Endocrinology, 17(10), 573-587.
  74. Stathi, D., et al. (2023). COVID-19 induced type 1 diabetes: A systematic review of case reports and series. Diabetologia, 66(1), 1–12. journals.sagepub.com
  75. D’Argenio, P., et al. (2021). SARS-CoV-2 and new-onset Type 1 diabetes in children. Acta Diabetologica, 58(2), 241-245.
  76. Kamrath, C., et al. (2021). New-onset type 1 diabetes in children and adolescents during the COVID-19 pandemic in Germany. Diabetes Care, 44(8), e163-e164.
  77. Wolf, S. E., et al. (2022). Increase in newly diagnosed type 1 diabetes in youth during the COVID‐19 pandemic in the United States: A multi‐center analysis. Pediatric Diabetes, 23(1), 38-46. onlinelibrary.wiley.com
  78. Cherubini, V., et al. (2021). New-onset type 1 diabetes in children and adolescents during the COVID-19 pandemic in Italy: an urgent need for an Italian observatory. Acta Diabetologica, 58(1), 115-117.
  79. Liu, F., et al. (2020). ACE2 expression in pancreatic β-cells and islet cells and its relevance to COVID-19. Endocrine, 68(1), 224-226.
  80. Eizirik, D. L., & Mandrup-Poulsen, T. (2001). A role for the interleukin-1β system in the pathogenesis of type 1 diabetes. Diabetologia, 44(Suppl 3), B106-B114.
  81. Mohan, V., & Unnikrishnan, R. (2021). COVID-19 and diabetes: The two-way street. Indian Journal of Endocrinology and Metabolism, 25(1), 1-2.
  82. American Diabetes Association. (2021). COVID-19 and Type 1 Diabetes: Addressing Concerns and Maintaining Control. Diabetes Care, 44(9), 1924–1926. diabetesjournals.org
  83. Singh, A. K., et al. (2020). Diabetes in COVID-19: prevalence, clinical course, prognosis and implications for care. Diabetes & Metabolic Syndrome: Clinical Research & Reviews, 14(6), 1973-1982.
  84. International Society for Pediatric and Adolescent Diabetes (ISPAD). (2020). COVID-19: Considerations for Children and Adolescents with Type 1 Diabetes. Journal of Diabetology, 2(3), 67-69. journals.lww.com
  85. Tinsley, L. J., et al. (2021). Rapid Transition to Telehealth for Adults with Type 1 Diabetes During the COVID-19 Pandemic. Diabetes Technology & Therapeutics, 23(1), 20-25.
  86. Hagger, M. S., et al. (2020). COVID-19, diabetes and mental health. Stress and Health, 36(5), 656-659.
  87. American Diabetes Association. (2024). Standards of Care in Diabetes—2024. Diabetes Care, 47(Suppl 1), S1-S302.

1 Comment

  1. Given the correlation observed between COVID-19 and new-onset T1D, could further research explore the potential for SARS-CoV-2 vaccination to act preventatively, mitigating the risk of T1D development in genetically predisposed individuals?

Leave a Reply to Brooke Mason Cancel reply

Your email address will not be published.


*