24
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Thirty Years of Research on the Dawn Phenomenon: Lessons to Optimize Blood Glucose Control in Diabetes

      article-commentary
      , MD, PHD, , MD, PHD, , MD, , MD, PHD
      Diabetes Care
      American Diabetes Association

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          More than 30 years ago in Diabetes Care, Schmidt et al. (1) defined “dawn phenomenon,” the night-to-morning elevation of blood glucose (BG) before and, to a larger extent, after breakfast in subjects with type 1 diabetes (T1D). Shortly after, a similar observation was made in type 2 diabetes (T2D) (2), and the physiology of glucose homeostasis at night was studied in normal, nondiabetic subjects (3–5). Ever since the first description, the dawn phenomenon has been studied extensively with at least 187 articles published as of today (6). In this issue, Monnier et al. (7) report an additional observation on the dawn phenomenon in a large group of T2D subjects and quantify its role on overall BG control. Given this information and the extensive data to date, an assessment of our knowledge in this area should be determined. Specifically, what have we learned from the last 30 years of research on the dawn phenomenon? What is the appropriate definition, the identified mechanism(s), the importance (if any), and the treatment of the dawn phenomenon in T1D and T2D? Physiology of glucose homeostasis in normal, nondiabetic subjects indicates that BG and plasma insulin concentrations remain remarkably flat and constant overnight, with a modest, transient increase in insulin secretion just before dawn (3,4) to restrain hepatic glucose production (4) and prevent hyperglycemia. Thus, normal subjects do not exhibit the dawn phenomenon sensu strictiori because they secrete insulin to prevent it. In T1D, the magnitude of BG elevation at dawn first reported was impressive and largely secondary to the decrease of plasma insulin concentration overnight (1), commonly observed with evening administration of NPH or lente insulins (8) (Fig. 1). Even in early studies with intravenous insulin by the “artificial pancreas” (Biostator) (2), plasma insulin decreased overnight because of progressive inactivation of insulin in the pump (9). This artifact exaggerated the dawn phenomenon, now defined as need for insulin to limit fasting hyperglycemia (2). When the overnight waning of insulin was prevented by continuous subcutaneous insulin infusion (CSII), even at single rate (10); intravenous infusion of albumin-added insulin by pump (11,12); or by the long-acting insulin analogs (LA-IAs) (8), it was possible to quantify the real magnitude of the dawn phenomenon—15–25 mg/dL BG elevation from nocturnal nadir to before breakfast (Fig. 1). Nocturnal spikes of growth hormone secretion are the most likely mechanism of the dawn phenomenon in T1D (13,14). The observation from early pioneering studies in T1D (10–12) that insulin sensitivity is higher after midnight until 3 a.m. as compared to the period 4–8 a.m., soon translated into use of more physiological replacement of basal insulin (CSII and the nearly peakless LA-IA [8] as compared with NPH) to reduce risk of nocturnal hypoglycemia while targeting fasting near-normoglycemia (Fig. 1). Figure 1 Overnight plasma insulin and glucose concentrations in normal, nondiabetic subjects (data from ref. 4) and in three groups of intensively treated subjects with T1D that had basal insulin replaced as either NPH (n = 6, NPH three times/day, at breakfast, lunch and bedtime; dose at bedtime 0.22 ± 0.03 units/kg/day), CSII (n = 6, infusion of basal insulin as rapid-acting analog at single rate 0.7 ± 0.1 U/h), or LA-IA (n = 6, dinnertime injection of glargine 0.25 ± 0.02 units/kg/day). Despite the fact that the total daily dose of NPH was fractionated into three times a day, bedtime NPH resulted in early plasma insulin peak and later insulin waning. This increases the risk of hypoglycemia after midnight and results in hyperglycemia before breakfast (dawn phenomenon 55 mg/dL). CSII at single rate prevents early peak and later overnight waning of insulin (dawn phenomenon 17 mg/dL). With LA-IA glargine, there is no dawn phenomenon since BG decreases in the second part of night as a result of continuing subcutaneous insulin absorption at dawn (7–8 h postinjection), which tends to slightly increase plasma insulin bioavailability at this time of day (G.B.B., unpublished observations). In T2D, identification of diurnal changes in BG goes back decades, but only quite recently fasting hyperglycemia has been attributed to a transient increase in hepatic glucose production (both glycogenolysis and gluconeogenesis) at dawn in the absence of compensatory insulin secretion (15–17). Monnier et al. (7) report on the overnight (interstitial) glucose concentration (IG), as measured by continuous ambulatory IG monitoring, in three groups of 248 subjects with T2D (on diet only, on insulin sensitizers alone, or on secretagogues alone or in combination with insulin sensitizers). They observed an increase in IG from nocturnal nadir to prebreakfast values similar in the three groups (13–20 mg/dL). The prebreakfast increase in IG extended to the postbreakfast period with the highest value of day (mean values 191–208 mg/dL). Importantly, the dawn phenomenon had an impact on mean daily IG and A1C (mean increase of 0.39% [4.3 mmol/mol]), which was independent of treatment. Two messages from the data of Monnier et al. (7) are important. First, the dawn phenomenon is confirmed as a frequent event across the heterogeneous population of T2D independent of (oral) treatment and studied in everyday life conditions, not only in the setting of specialized clinical research units. Second, the article reaffirms that the primary target of treatment in T2D is to reestablish near-normoglycemia before and after breakfast (i.e., to treat the dawn phenomenon) to lower mean daily BG and A1C (8). The absolute overnight increase in fasting IG observed (7) is in the range of that reported previously for BG at dawn (10–12,17), and the postbreakfast IG is higher as compared with the postlunch and dinner values (7). Thus, the dawn phenomenon induces hyperglycemia not only before, but, to a larger extent, after breakfast as well (7,18). Over the years, fasting (and postbreakfast) hyperglycemia in T2D worsens as result of progressively impaired pancreatic B-cell function on the background of continued insulin resistance primarily at dawn (8,15–18) and independently of age (19). Because it is an early metabolic abnormality leading over time to the vicious circle of “hyperglycemia begets hyperglycemia” by glucotoxicity and lipotoxicity, the dawn phenomenon in T2D should be treated early and appropriately before A1C continues to increase (20). Oral medications do not adequately control the dawn phenomenon, even when given in combination (7,18). Sulphonylureas are less than ideal due to risk of hypoglycemia in the afternoon or evening, when the dose is increased to counteract the hyperglycemia of dawn phenomenon. Incretins are designed to elegantly improve the postprandial, not the fasting, periods (20). The evening replacement of basal insulin, which abolishes the dawn phenomenon by restraining hepatic glucose production and lipolysis (21), is an effective treatment as it mimics the physiology of glucose homeostasis in normal, nondiabetic subjects (4). Early use of basal insulin in T2D is an add-on option treatment after failure of metformin to control A1C <7.0% (20). However, to fight the clinical inertia, it would be wise to consider initiation of basal insulin (effective, easy, safe, and “natural”) before—not after—A1C has increased well beyond 7.0%, as usually it is done in practice currently. Basal insulin may be initiated to prevent the increase of A1C >7.0% (22) by reducing nocturnal hyperglycemia and the dawn phenomenon (Fig. 2). Figure 2 Overnight plasma glucose and insulin concentrations in a group of T2D subjects (n = 8, age 53 ± 4 years, diabetes duration 3 ± 1 years, A1C 6.89 ± 0.05% [52 ± 0.5 mmol/mol], all on metformin only) before and after 6-month treatment with evening dose of insulin glargine (0.20 ± 0.02 U/Kg/day) as add-on to metformin. Basal insulin near-normalized the fasting BG by two mechanisms: partly by reducing the midnight BG and partly by totally abolishing the BG increase of the dawn phenomenon of the baseline study (18 mg/dL), as result of overnight sustained increase in plasma insulin concentration by ∼4 μU/mL. At the end of observation, the removal of the dawn phenomenon resulted in a decrease of A1C of 6.5 ± 0.1% (48 ± 0.7 mmol/mol). This validates the estimated contribution of the dawn phenomenon to the increased A1C calculated by Monnier et al. (7) of 0.39% (4.3 mmol/mol) (G.B.B., unpublished data). In summary, despite the extensive number of publications today, the dawn phenomenon remains an area of research interest and, importantly, a target for clinical modulation that should be taken into account whenever choosing treatments.

          Related collections

          Most cited references18

          • Record: found
          • Abstract: found
          • Article: not found

          The loss of postprandial glycemic control precedes stepwise deterioration of fasting with worsening diabetes.

          The aim of the study was to determine whether the loss of fasting and postprandial glycemic control occurs in parallel or sequentially in the evolution of type 2 diabetes. In 130 type 2 diabetic patients, 24-h glucose profiles were obtained using a continuous glucose monitoring system. The individuals with type 2 diabetes were divided into five groups according to A1C levels: 1 ( or =9%, n = 26). The glucose profiles between the groups were compared. The overall glucose concentrations for the diurnal, nocturnal, and morning periods, which represent the postprandial, fasting, and the dawn phenomenon states, respectively, were also compared. Glucose concentrations increased steadily from group 1 to 5 in a stepwise manner. The initial differences in mean glucose concentrations reaching statistical significance occurred 1) between groups 1 and 2 (6.4 vs. 7.7 mmol/l, P = 0.0004) for daytime postprandial periods, followed by differences; 2) between groups 2 and 3 (7.5 vs. 9.3 mmol/l, P = 0.0003) for the morning periods (dawn phenomenon); and finally 3) between groups 3 and 4 (6.3 vs. 8.4 mmol/l, P < 0.0001) for nocturnal fasting periods. The deterioration of glucose homeostasis in individuals with type 2 diabetes progressed from postprandial to fasting hyperglycemia following a three-step process. The first step related to the three diurnal postmeal periods considered as a whole, the second step occurred during the morning period, and the third and final step corresponded to sustained hyperglycemia over the nocturnal fasting periods. Such a description of the key stages in the evolution of type 2 diabetes may be of interest for implementing antidiabetes treatment.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Evidence for a circadian rhythm of insulin sensitivity in patients with NIDDM caused by cyclic changes in hepatic glucose production.

            Diurnal variation in insulin sensitivity in patients with NIDDM has long been suspected but has been difficult to document mainly because of the interdependence of changes in glucose and insulin. Stable serum insulin levels during hyperglycemic clamping in patients with NIDDM in the present study provided the opportunity to examine changes in insulin sensitivity unaffected by changes in blood glucose and insulin concentrations. Six patients with NIDDM (four men and two women, BMI 33.9 +/- 2.5) underwent hyperglycemic (11.1 mmol/l, approximately 200 mg/dl) clamping for 72 h. Measured were serum insulin, free fatty acid (FFA), cortisol, and growth hormone concentrations and rates of insulin secretion, insulin clearance, and glucose infusion rate (GIR) needed to maintain hyperglycemia. In addition, five patients (three men and two women, BMI 32.6 +/- 0.6) underwent hyperglycemic clamping for 24 h with hourly determinations of hepatic glucose production (HGP) and glucose disappearance rates (GRd). GIR, reflecting insulin sensitivity, changed rhythmically with a cycle duration of 22.9 +/- 1.4 h and an amplitude of 47.8 +/- 11.2%. GIR was lowest at 8:31 a.m. (+/- 52 min) and highest at 7:04 p.m. (+/- 58 min). Circadian changes in GIR were completely accounted for by changes in HGP, while GRd remained unchanged. Plasma levels of FFAs and cortisol also exhibited circadian fluctuations, and their blood levels correlated negatively with GIR (r = -0.72 and -0.64, respectively). We concluded that insulin sensitivity in patients with NIDDM changed with circadian (approximately 24 h) rhythmicity (decreasing during the night and increasing during the day). These changes were unrelated to blood levels of glucose and insulin, insulin clearance, exercise, food intake, and sleep. They were caused by circadian changes in HGP, which in turn were closely correlated with circadian changes in blood FFA and cortisol levels. We believe that recognition of these circadian changes has implications for the diagnosis and the treatment of patients with NIDDM.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              The "dawn phenomenon"--a common occurrence in both non-insulin-dependent and insulin-dependent diabetes mellitus.

              The dawn phenomenon is a condition recently described in patients with insulin-dependent diabetes mellitus (IDDM) that is characterized by abrupt increases in fasting levels of plasma glucose or insulin requirements or both between 5 and 9 a.m., in the absence of antecedent hypoglycemia. To determine its potential clinical relevance, we assessed its frequency and reproducibility in 20 patients with IDDM and in 13 patients with non-insulin-dependent diabetes mellitus (NIDDM) during overnight closed-loop (feedback-controlled) intravenous insulin infusion. After 6 a.m., plasma glucose levels increased similarly in NIDDM (89 +/- 2 mg per deciliter, midnight to 6 a.m., vs. 98 +/- 2 mg per deciliter, 6 to 9 a.m.; P less than 0.01). Insulin requirements increased at least 50 per cent for 1 1/2 hours in 77 per cent of patients with NIDDM and in 75 per cent of patients with IDDM. In five patients with IDDM who were studied on four occasions, the phenomenon occurred during 17 of the 20 observation periods, with insulin requirements after 6 a.m. increasing 225 +/- 34 per cent; coefficients of variation in individual patients ranged from 4 to 25 per cent. Thus, the dawn phenomenon occurs commonly in both NIDDM and IDDM, but its potential variability must be taken into consideration when one is attempting to adjust insulin doses.
                Bookmark

                Author and article information

                Journal
                Diabetes Care
                Diabetes Care
                diacare
                dcare
                Diabetes Care
                Diabetes Care
                American Diabetes Association
                0149-5992
                1935-5548
                December 2013
                13 November 2013
                : 36
                : 12
                : 3860-3862
                Affiliations
                [1]Department of Medicine, University of Perugia, Perugia, Italy
                Author notes
                Corresponding author: Geremia B. Bolli, geremia.bolli@ 123456unipg.it .
                Article
                2088
                10.2337/dc13-2088
                3836156
                24265365
                72566caf-cadc-456b-80e5-37c7ef0d9183
                © 2013 by the American Diabetes Association.

                Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.

                History
                Page count
                Pages: 3
                Categories
                Commentaries

                Endocrinology & Diabetes
                Endocrinology & Diabetes

                Comments

                Comment on this article