Abstract
Sodium-glucose cotransporter-2 (SGLT2) inhibition and lactation result in the excretion of large amounts of glucose in urine or milk and are associated with a lower risk of cardiovascular events. The respective mechanisms behind this association with cardiovascular protection are not clear. This review compares the contribution of noninsulin-mediated glucose transport during pharmacologic inhibition of SGLT2 with noninsulin-mediated glucose transport during lactation in terms of the implications for the cardiometabolic health of parous women. The search topics used to obtain information on SGLT2 inhibitors included mechanisms of action, atherosclerosis, and heart failure. The search topics used to obtain information on lactation included cardiovascular health and milk composition. Subsequent reference searches of retrieved articles were also used. Active treatment with SGLT2 inhibitors affects glucose and sodium transport in the kidneys and predominantly protects against hospitalization for heart failure soon after the onset of therapy. Active lactation stimulates glucose transport into the mammary gland and improves subclinical and clinical atherosclerotic vascular disease years after delivery. Both SGLT2 inhibitors and lactation have effects on a variety of glucose transporters. Several mechanisms have been proposed to explain the cardiometabolic benefits of SGLT2 inhibition and lactation. Learning from the similarities and differences between both processes will advance our understanding of cardiometabolic health for all people.
Keywords: breastfeeding, cardiovascular, GLUT1, lactation, postpartum, SGLT2
Introduction
Excretion of glucose in body fluids like urine and milk reduces blood glucose concentrations and induces systemic effects that alter metabolism in distant organs. Increased glucose excretion can be achieved by pharmacologic or physiological means while the stimulus lasts. This stimulus can be a drug like a sodium-glucose cotransporter-2 (SGLT2) inhibitor or a physiological state like lactation. In view of the undisputed benefits of SGLT2 inhibitors on cardiovascular health and the consistent observations that lactation decreases maternal cardiovascular risk, this review compares SGLT2 inhibition and lactation as strategies that promote multiorgan protection beyond their glucose-lowering effects.
Slowing down the atherosclerosis process would likely require years of sustained improvement in risk factors for atherosclerotic cardiovascular disease (ASCVD).1 SGLT2 inhibitors significantly reduce hospitalization for heart failure, atherosclerotic events, cardiovascular, and total mortality as well as progression of chronic kidney disease.2,3 However, the reductions in adverse cardiovascular outcomes and hospitalizations for heart failure observed in clinical trials had an earlier onset than anticipated by an effect in reducing atherosclerosis. In comparison, the lifetime history of lactation combines relatively small but sustained improvements in cardiometabolic risk factors with fewer cardiovascular events.4
Glucose Transport Mediated by SGLT2: Metabolic Effects of SGLT2 Inhibition
SGLT2 is a glucose sensor that transports glucose and sodium from the tubular lumen into the proximal tubule cells of the kidney independently of insulin.5 SGLT2 inhibitors block the SGLT2 transporter resulting in glucosuria of 50–100 grams per day depending on blood glucose concentration and drug dose.6,7 This amount of glucosuria decreases both glucotoxicity and the flux of glucose through the hexosamine biosynthesis pathway, improving both insulin secretion during hyperglycemic clamps and insulin sensitivity during hyperinsulinemic-euglycemic clamps.8,9 SGLT2 inhibitors reduce fasting glucose and insulin concentrations, characteristics that are associated with health and longevity in other contexts.10 SGLT2 inhibitors also increase endogenous glucose production (EGP) in the fasting state and promote lipid oxidation.11 This drug class may reduce body weight, visceral fat, lipotoxicity, and hepatic steatosis.2,12 These pharmacologic effects have a rapid onset and only persist during active treatment. SGLT2 inhibitors induce a fasting-like state characterized by increased nutrient deprivation signaling and autophagic flux, and a decrease in nutrient surplus signaling, apoptosis, and senescence.13 The heart does not express SGLT2 transporters.14
SGLT2 Inhibitors: Mechanisms of Protection
Multiple mechanisms have been proposed to explain the organ-protective effects of SGLT2 inhibitors. These mechanisms include the impact of SGLT2 inhibitors on metabolism and hemodynamics.15 Specific examples are inducing a fasting-like state with shifts in energy metabolism from glucose to fat oxidation, modification of cellular life history programs, increase in hematocrit, natriuresis, decrease in blood pressure and inflammation, effects on plaque burden, size, and stability, and nonatherosclerotic mechanisms.2,16–20 Aggregated clinical trials data suggest a potential benefit of SGLT2 inhibitors on myocardial infarction beyond the consistent findings of improvement in ejection fraction among patients with heart failure.21
The benefits of SGLT2 inhibition occur in different contexts. Some benefits depend on glucosuria or the interaction between the SGLT2 inhibitor drug and the SGLT2 transporter.14,22,23 Increased glucosuria promotes a glucose deficit, lowers glucose and insulin in circulation, disinhibits lipolysis, increases circulating free fatty acids (FFAs) and ketone bodies, and promotes a switch in substrate utilization.22 Selective inhibition of SGLT2-mediated glucose and sodium reabsorption in the proximal tubule of the kidneys, leads to the complement of glucosuria, natriuresis, and osmotic diuresis. These changes promote a reduction in intravascular volume, arterial blood pressure, cardiac preload, and cardiac afterload.2,24 Among subjects with the triad of type 2 diabetes, chronic kidney disease, and no known preexisting ASCVD, those who had the highest glucosuria while treated with the SGLT2 inhibitor canagliflozin, also had the strongest protection against multiple cardiorenal outcomes.22
SGLT2 inhibitors also have effects that are independent of glucosuria. The finding that SGLT2 inhibition still confers cardiac protection in patients with low glomerular filtration rate and relatively low glucosuria and the findings in experimental models with isolated cardiomyocytes suggest that this drug class also has a direct effect on the heart.5,11,14,23 A study with molecular docking analysis with the SGLT2 inhibitor empagliflozin showed that empagliflozin directly binds three glucose transporters.14 Empagliflozin had a higher affinity for glucose transporter 1 (GLUT1) and GLUT4 compared with SGLT1. In addition, the SGLT2 inhibitor canagliflozin directly binds mammalian target of rapamycin (mTOR) in cardiomyocytes and inhibits mTOR phosphorylation.23 It is not clear if the mechanisms of protection are related to kidney-related systemic alterations, direct cardiovascular effects, or both.25
Targeted metabolomics of plasma samples in a placebo-controlled trial of dapagliflozin in patients with heart failure with reduced ejection fraction supported a role for altered ketone and fatty acid biology with SGLT2 inhibitors.26 This study showed that dapagliflozin increased ketone body and FFA-related metabolites, which suggested augmented consumption of both ketone bodies and FFAs in the myocardium by SGLT2 inhibitors.27 However, no change in cardiac energetics or targeted circulating metabolites associated with energy metabolism were found in a mechanistic cardiac MR study.28 This mechanistic study was designed to investigate the effects of treatment with empagliflozin on cardiac physiology and metabolism in patients with heart failure.28
Lactation: Glucose Transport and Maternal Metabolism
Lactation is one of the most metabolically challenging events in the lifetime of a woman when the body prioritizes glucose transport to the mammary gland over glucose transport to most other tissues in the body.29 Lactation is thought to rely on the facilitated GLUT1 to transport 50–75 grams of glucose per day into the mammary epithelial cells (MECs) for the production of milk components including lactose and other carbohydrates, lipids, and proteins.30–33 The concentration of free glucose in milk is very low and resembles the concentration of free glucose in the cytoplasm of MECs.34,35 The amount of glucose transported into milk varies by blood glucose concentrations and by the intensity of the lactation stimulus (proportion of milk fed to the infant as mother’s milk relative to formula).32,35
GLUT1 is responsible for basal glucose transport without the mediation of insulin. Besides its ubiquitous location in adults, GLUT1 is the dominant glucose transporter in cardiomyocytes, skeletal myocytes, and liver during fetal life.36,37 During lactation and weaning, the proportion of GLUT1 in the tissues of the offspring decreases relative to other facilitated glucose transporters.36 This suggests the possibility that reliance of maternal tissues on GLUT1 during lactation resets maternal glucose uptake to an earlier program, adaptive to the uniqueness of the postpartum period. GLUT1 can transport glucose and galactose, the two monosaccharides needed to make the disaccharide lactose.38,39
At least three different membrane compartments participate in the transport of glucose in MECs: (1) the basolateral membrane, which is the side of the plasma membrane more proximal to maternal circulation; (2) the Golgi apparatus where synthesis of lactose takes place; and (3) the apical membrane, which is the side of the plasma membrane closest to milk.38 Most of what is known about glucose transport in MECs comes from studies with animals and the level of detail addressed in animal studies is impossible to attain with human studies. GLUT1 is the predominant (but not exclusive) glucose transporter in the basolateral membrane of MECs in rats, mice, and cows.38,40 A steep glucose concentration gradient drives glucose transport across the basolateral membranes of MECs.
In humans, the expression of a variety of glucose transporters has been identified in MECs or milk fat globules obtained from milk samples.39,41–43 Obermeier et al. isolated cells from fresh human milk at or after 1 month postpartum.39 These investigators detected SGLT1 mRNA and protein expression without indication of GLUT1 mRNA expression. SGLT1 like GLUT1 can transport glucose and galactose.39 This study was not designed to determine the membrane compartment(s) where SGLT1 localizes. One interpretation is that SGLT1 transport is more relevant at the apical membrane because the glucose concentration inside the MECs resembles the glucose concentration in milk. SGLT2 and GLUT4 (GLUT4 is the transporter tasked with insulin-mediated glucose uptake) have not been detected in MECs. Glucose transporters other than GLUT1/GLUT4 and SGLT1/SGLT2 have been detected by single-cell RNA sequencing of human milk-derived cell populations during early lactation and by studies on mammary cell RNA from human milk fat globules.41,42
In lactating women, increasing the glucose concentration in plasma resulted in a two- to three-fold increase in the glucose concentration in milk, consistent with facilitated diffusion of glucose across the basolateral membrane.32 Serum insulin concentrations between 2 and 70 µU/mL have no effect on human milk glucose.32 This glucose uptake independent of insulin concentrations is consistent with the lack of GLUT4 in MECs.43,44 GLUT1 and GLUT4 are the main glucose transporters in skeletal muscle and adipose tissue, the two main tissues controlling whole-body insulin sensitivity.45–47
The body of evidence supports that multiple glucose transporters contribute to moving glucose across the plasma membrane and organelle compartments of the MECs.38–42 In humans, the functional aspects of glucose transport are consistent with the predominance of GLUT1 transport in the basolateral membrane.32 Other glucose transporters likely contribute to the movement of glucose across MECs but in less proportion. Once glucose crosses the basolateral membrane, it is lost to maternal circulation and the interstitial space.38
Lastly, lactation reduces fasting glucose and insulin concentrations, characteristics that are associated with health and longevity in other contexts.10,48,49 Low insulin could promote nutrient deprivation signaling and autophagic flux. Lactation increases EGP in the fasting state and may reduce visceral fat, lipotoxicity, and hepatic steatosis.50,51 The protective association persists after adjusting for changes in body weight.
Lactation: Mechanisms of Protection
In humans, little is known about metabolic adaptations during active lactation and postlactation responsible for about 50% reduction in the long-term risk of type 2 diabetes and metabolic syndrome in the mother.30,52,53 Viewing the lactating mammary gland as a sink for glucose and calories provides the simplest explanation for the immediate health benefits of active lactation on maternal metabolism. However, the leap from active benefits to lasting protection does not have a clear explanation yet.
Observational studies and studies with mice support the reset hypothesis, which states that lactation promotes recovery from the increased cardiometabolic stresses of normal pregnancy closer to the prepregnancy state.54–56 These pregnancy stresses include accumulation of visceral fat, hyperlipidemia, and decreased insulin sensitivity.57,58 The variable duration and intensity of lactation after each pregnancy result in a cumulative effect over cycles of pregnancy-lactation throughout the reproductive years. A longer lifetime duration of lactation is associated with a reduction of several maternal cardiovascular risk factors including type 2 diabetes, hypertension, hyperlipidemia, obesity, and metabolic syndrome after adjusting for other healthy behaviors.30,53,59 In addition, a longer duration of lactation appears to benefit the coagulation profile, particularly among women with a history of gestational diabetes.60 Slowing down the atherosclerosis process would likely require years of sustained improvement in risk factors for ASCVD.1 Importantly, the beneficial effect of lactation on cardiometabolic risk can persist for 15 years after the last birth.30,59
In this regard, the Coronary Artery Risk Development in Young Adults study showed that longer duration of lactation was associated with both a lower incidence of the metabolic syndrome and type 2 diabetes among parous women with and without a history of gestational diabetes, years after cessation of lactation.30,53 This association was present even after adjustment for weight change in the follow-up period. Lactation also reduces the risk of subclinical and clinical cardiovascular disease.61–64 Several studies have shown an association between lactation and improvements in the vascular health of the carotids, coronaries, and aorta.61,63
Among parous women in the Nurses’ Health Study, a lifetime duration of lactation of 2 years or longer was associated with a decreased risk of incident myocardial infarction compared with women who had never lactated.64 In addition, a recent meta-analysis that included eight observational studies and over a million pregnancies documented that any duration of lactation was associated with an 11%−17% reduction in the relative risk of cardiovascular disease, coronary heart disease, stroke, and fatal cardiovascular disease.65 To date, there are no published clinical studies on the effect of lactation on the prevention of heart failure. However, translational work with mice has shown increased ejection fraction as a benefit of lactation versus no lactation.55,56 This increase in ejection fraction was already present at 2 months postpartum (after weaning) and extended into the mouse midlife at 9 months postpartum.
Effects of SGLT2 Inhibition and Lactation on Weight and Body Composition
SGLT2 inhibition and lactation share several characteristics that result in changes in body weight and composition. These changes include loss of energy in a body fluid (urine or milk) and a compensatory increase in caloric intake that result in lower-than-predicted weight loss.66–68 Both SGLT2 inhibition and lactation decrease waist circumference, percent body fat, visceral, subcutaneous, and hepatic fat.50,51,69–72 However, their effect on lean mass and skeletal muscle mass is variable.69,73
The glucosuria induced by SGLT2 inhibitors (50–100 grams/day) has an equivalent loss of energy in the urine of 200–400 kcal/day.7 This energy loss triggers an increase in caloric intake that limits weight loss to 2–4 kg.7 Two-thirds of the weight loss is fat mass and one-third is fat-free mass.66 SGLT2 inhibitors induce weight loss in patients with and without diabetes and decrease fat in soft tissue compartments: visceral, subcutaneous, hepatic, and epicardial.70,71,74,75 This drug class also decreases lean and skeletal muscle mass.69
The trajectories of weight and body composition during lactation are influenced by the trajectories of weight and body composition during pregnancy.58 Weight gain during pregnancy impacts the patterns of postpartum weight retention between lactating and nonlactating women partly because weight gain during pregnancy is the major determinant of postpartum weight retention.76 Although the direction and magnitude of weight change during lactation varies, women who lactate during the first year postpartum lose on average 2 kg more than formula-feeding women from 1 to 12 months postpartum.67 Women also experience an increase in visceral fat during pregnancy.58 Longer duration of lactation is associated with lower visceral fat volumes before and after menopause as well as a lower risk of metabolic dysfunction-associated steatotic liver disease in midlife.50,51,77
Differences Between SGLT2 Inhibition and Lactation
SGLT2 inhibitors and lactation have important differences regarding the onset and duration of alterations in metabolism, target organs that reap protective effects, the magnitude of the starvation stimulus, and the effect on insulin secretion and insulin sensitivity. The benefit obtained during active treatment with SGLT2 inhibitors appears to cease with discontinuation of therapy. Treatment with SGLT2 inhibitors induces an increase in glucagon concentration and fat oxidation as well as the development of ketosis in a sort of accelerated starvation.6 SGLT2 inhibitors improve insulin secretion when adjusting for glucose during a hyperglycemic clamp.8 SGLT2 inhibitors improve insulin sensitivity during a hyperinsulinemic-euglycemic clamp after correction for urinary loss of glucose.9
The pharmacologic inhibition of SGLT2 in the proximal renal tubule and the stimulation of nutrient transport into the lactating mammary gland result in loss of calories in their respective body fluids, urine, or milk. The amount of glucose excreted in urine in response to SGLT2 inhibition increases in proportion to the plasma glucose concentration.5 In contrast, women who only feed their infants with mother’s milk produce an average 750–800 mL of milk per day once lactation is established.31,34,78,79 Considering an energy content of 65–70 kcal per 100 mL of milk, the amount of energy in milk ranges from 480 to 560 kcal/day with a mixed macronutrient composition that includes fat, carbohydrate, and protein.80 About 55% of this energy is fat, 40% is carbohydrate, and 5% is protein.32,81,82
Lactation is associated with cardiometabolic benefits long after weaning.30,53 Compared with women who only feed formula, lactating women have lower glucagon concentrations at 3 and 6 months postpartum, suppression of lipolysis at lower insulin concentrations, and onset of ketosis following a 24–42-hr fast rather than during an overnight fast.31,49,83 These characteristics suggest that compared with SGLT2 inhibition, the starvation stimulus during lactation is gentler on nonmammary tissues. Although lactating women exhibit similar or decreased insulin secretion during a 2-hr oral glucose tolerance test (OGTT) or during an intravenous glucose tolerance test (IVGTT) when compared with formula-feeding women, lactating women also exhibit an improved disposition index during an IVGTT.84–88 These changes suggest that the insulin secretory response is appropriate during lactation.
Specific to insulin sensitivity, longitudinal assessments between pregnancy and 3 years postpartum using OGTT showed better insulin sensitivity by Matsuda Index in women who lactated 12 months or longer compared with women who lactated 3 months or less and in women with a history of lactation versus no lactation.84,89 Published work with hyperinsulinemic-euglycemic clamps during lactation has not accounted for the loss of glucose into milk during calculations of insulin-stimulated glucose disposal.35,85 A large proportion of glucose uptake during lactation appears to be noninsulin-mediated glucose uptake, typically associated with GLUT1 transport. Therefore, adaptations in glucose transport may confound the interpretation of whole-body and tissue-specific sensitivity to insulin among lactating women. Figure 1 compares the important effects of SGLT2 inhibitors and lactation on metabolism.
FIG. 1.
Effect of SGLT2 inhibitors and lactation on the main tissues involved in glucose metabolism. (A) SGLT2 inhibitors induce excretion of 50–100 grams of glucose per day in urine. This drug class promotes a systemic response that mimics starvation despite also inducing hyperphagia. The responses in major metabolic tissues include decreases in hepatic steatosis, visceral and subcutaneous fat, and a mean weight loss of 2–4 kg; and increases in endogenous glucose production (EGP), lipid oxidation, and insulin sensitivity. Insulin secretion improves during hyperglycemic clamps. (B) Active lactation induces excretion of 50–75 grams of glucose per day in milk and is associated with hyperphagia. The responses in major metabolic tissues include decreases in hepatic steatosis, visceral and subcutaneous fat, and increases in EGP. It is unclear how insulin-sensitive are the nonmammary tissues of lactating women. It is not known if glucose uptake in nonmammary tissues is mainly dependent or mainly independent of insulin. Relative to formula-feeding women, insulin secretion decreases during lactation while the amount of secreted insulin for the magnitude of insulin resistance increases.
Differential Effects of SGLT2 Inhibition and Lactation on Glucagon and Incretin Hormones
Glucagon potentiates glucose-stimulated insulin secretion (GSIS), stimulates energy expenditure and hepatic glucose production (HGP), and decreases glucose effectiveness.90–93 Glucose effectiveness refers to the ability of glucose itself to suppress EGP and stimulate peripheral glucose uptake as noninsulin-mediated glucose uptake.93 Glucagon-like peptide 1 (GLP-1) potentiates GSIS, induces satiety, decreases food intake, inhibits HGP, and increases glucose effectiveness.91,93–95 Glucose-dependent insulinotropic polypeptide also potentiates GSIS and increases glucose effectiveness.93,95
Specific to SGLT2 inhibition, the alpha cells in human islets express SLC5A2 mRNA (encoding SGLT2).96 The expression of SLC5A2 as well as glucagon secretion exhibit marked interindividual variation, which may contribute to the range of glucagon responses to SGLT2 inhibitors in patients with diabetes.96 The metabolic effects of SGLT2 inhibitors vary along the spectrum of glucose tolerance.6 In the context of normal glucose tolerance (NGT), treatment with SGLT2 inhibitors did not affect fasting plasma glucose, insulin, or glucagon concentrations despite glucosuria of about 50 grams/day.6,97 Individuals with NGT had stable mean fasting plasma glucagon concentrations ranging from 69 to 75 pg/mL during the 14-day period of study before and after the introduction of SGLT2 inhibition. Furthermore, adults with obesity and without diabetes had similar plasma glucagon when treated with SGLT2 inhibitors or placebo.71 The intermediate group with impaired fasting glucose had a small but significant decrease in fasting plasma glucose with no change in fasting plasma insulin or glucagon.6 Lastly, adults with type 2 diabetes had a significant decrease in fasting plasma glucose and a significant increase in fasting plasma glucagon within 1 day of the introduction of SGLT2 inhibition.6
Two studies comparing fasting plasma glucagon in lactating versus nonpregnant, nonlactating women reported similar glucagon concentrations between the groups (∼76 pg/mL).31,98 A longitudinal evaluation of well-nourished women followed between term pregnancy and 6 months postpartum, reported higher mean glucagon concentrations during pregnancy (∼160 pg/mL) compared with the postpartum period and higher serum glucagon in nonlactating (∼138 pg/mL) compared with lactating women at 3 months (81 pg/mL) and at 6 months postpartum (83 pg/mL).49 These three independent studies reported similar fasting glucagon concentrations among lactating women.31,49,98 A fourth study on this subject reported fasting and postprandial concentrations of GLP-1 at 4–6 weeks postpartum in women who only fed their infants with mother’s milk versus a never-pregnant control group. This study found no differences in GLP-1 concentrations between the groups, which were matched for age and body composition.99
Lactation is characterized by low insulin concentrations, increased EGP in the fasting state, increased appetite, and presumed increased glucose effectiveness because of the reliance of a large proportion of glucose utilization on noninsulin-mediated glucose transport to the MECs for milk production. The contribution of the vast concurrent and sequential hormonal changes that occur during pregnancy and the postpartum period to these processes once lactation is established is not clear.
Implications for Cardiometabolic Health in Parous Women
SGLT2 inhibition and lactation result in the excretion of large amounts of glucose in urine or milk and are associated with a lower risk of cardiovascular events. The mechanisms behind the association of SGLT2 inhibition with cardiovascular protection are not clear. In whole organisms, it is difficult to separate the systemic contribution of glucosuria from direct cardiovascular effects when SGLT2 inhibitors bind to molecules other than the SGLT2 transporter, which is not expressed in cardiomyocytes. Likewise, the mechanisms behind the association of lactation with cardiovascular protection are not clear. The reset hypothesis links relatively small but sustained improvements in a cluster of cardiometabolic risk factors (blood pressure, lipids, glycemia, body weight, and body composition) with fewer cardiovascular events.54 The reset hypothesis is a compelling argument when considering that slowing down the atherosclerosis process would likely require years of sustained improvement in its risk factors.1
In general terms, the systemic effects observed with SGLT2 inhibitors and lactation result from the combination of tissue-specific effects and inter-tissue communication. SGLT2 inhibition and lactation involve glucose transport. SGLT2 inhibition and lactation also affect cardiometabolic risk. Despite similarities in many metabolic effects, their specific mechanisms of protection may differ. Other potential mechanisms for improvement in metabolic health among parous women are lactation and postlactation effects on bone health and stem cells.100 The duration of postlactation recovery can be defined as the 6 to 12 months that it takes the maternal skeleton to recover from the bone loss induced by lactation.101 Such recovery implies prolonged changes in the bioenergetics of bone to build back to the prepregnancy state.
In terms of stem cells, Avogaro et al. suggested that the energy deficit with SGLT2 inhibitors induces a dormancy program linked to autophagy and stem cell quiescence.17 Skeletal stem and progenitor cells (SSPC) are known to rejuvenate in contexts other than the pregnancy-postpartum periods under conditions of energy deficit that upregulate autophagy.102–104 Avogaro et al. asked, “Can the effect of SGLT2 inhibitors on cellular reprogramming occur with other noninsulin-dependent reductions in plasma glucose concentration?”17 If cellular reprogramming occurred during lactation, would it be possible for the rejuvenation of SSPC to partly explain the durable cardiometabolic benefit associated with lactation? Learning from the similarities and differences between SGLT2 inhibitors and lactation will advance our understanding of cardiometabolic health for all people. Clinical research on the physiology of lactation and the postlactation period is critically needed to promote the health of parous women during their reproductive years. Only then will the mechanisms behind this protective association be identified.
Author Disclosure Statement
The author declares no conflict of interest.
Funding Information
The ideas in this review originated during the period of funding by the National Institutes of Health grant R03DK097463 (MARR) and by the American Diabetes Association grant 1-14-TS-33 (MARR).
References
- 1. Sattar N, Deanfield J, Delles C. Impact of intentional weight loss in cardiometabolic disease: What we know about timing of benefits on differing outcomes? Cardiovasc Res 2024;119(18):e167–e171; doi: 10.1093/cvr/cvac186 [DOI] [PubMed] [Google Scholar]
- 2. Zelniker TA, Braunwald E. Mechanisms of cardiorenal effects of sodium-glucose cotransporter 2 inhibitors: JACC state-of-the-art review. J Am Coll Cardiol 2020;75(4):422–434; doi: 10.1016/j.jacc.2019.11.031 [DOI] [PubMed] [Google Scholar]
- 3. He G, Yang G, Huang X, et al. SGLT2 inhibitors for prevention of primary and secondary cardiovascular outcomes: A meta-analysis of randomized controlled trials. Heart Lung 2023;59:109–116; doi: 10.1016/j.hrtlng.2023.02.009 [DOI] [PubMed] [Google Scholar]
- 4. Kim C, Catov J, Schreiner PJ, et al. Women’s reproductive milestones and cardiovascular disease risk: A review of reports and opportunities from the CARDIA study. J Am Heart Assoc 2023;12(5):e028132; doi: 10.1161/JAHA.122.028132 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. DeFronzo RA, Reeves WB, Awad AS. Pathophysiology of diabetic kidney disease: Impact of SGLT2 inhibitors. Nat Rev Nephrol 2021;17(5):319–334; doi: 10.1038/s41581-021-00393-8 [DOI] [PubMed] [Google Scholar]
- 6. Al Jobori H, Daniele G, Adams J, et al. Determinants of the increase in ketone concentration during SGLT2 inhibition in NGT, IFG and T2DM patients. Diabetes Obes Metab 2017;19(6):809–813; doi: 10.1111/dom.12881 [DOI] [PubMed] [Google Scholar]
- 7. Ferrannini E, Muscelli E, Frascerra S, et al. Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients. J Clin Invest 2014;124(2):499–508; doi: 10.1172/JCI72227 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Merovci A, Mari A, Solis-Herrera C, et al. Dapagliflozin lowers plasma glucose concentration and improves β-cell function. J Clin Endocrinol Metab 2015;100(5):1927–1932; doi: 10.1210/jc.2014-3472 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Merovci A, Solis-Herrera C, Daniele G, et al. Dapagliflozin improves muscle insulin sensitivity but enhances endogenous glucose production. J Clin Invest 2014;124(2):509–514; doi: 10.1172/JCI70704 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Kenyon CJ. The genetics of ageing. Nature 2010;464(7288):504–512; doi: 10.1038/nature08980 [DOI] [PubMed] [Google Scholar]
- 11. Ferrannini E. Sodium-glucose co-transporters and their inhibition: Clinical physiology. Cell Metab 2017;26(1):27–38; doi: 10.1016/j.cmet.2017.04.011 [DOI] [PubMed] [Google Scholar]
- 12. Packer M. Role of deranged energy deprivation signaling in the pathogenesis of cardiac and renal disease in states of perceived nutrient overabundance. Circulation 2020;141(25):2095–2105; doi: 10.1161/CIRCULATIONAHA.119.045561 [DOI] [PubMed] [Google Scholar]
- 13. Packer M. Critical reanalysis of the mechanisms underlying the cardiorenal benefits of SGLT2 inhibitors and reaffirmation of the nutrient deprivation signaling/autophagy hypothesis. Circulation 2022;146(18):1383–1405; doi: 10.1161/CIRCULATIONAHA.122.061732 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Li X, Lu Q, Qiu Y, et al. Direct cardiac actions of the sodium glucose co-transporter 2 inhibitor empagliflozin improve myocardial oxidative phosphorylation and attenuate pressure-overload heart failure. J Am Heart Assoc 2021;10(6):e018298; doi: 10.1161/JAHA.120.018298 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Panico C, Bonora B, Camera A, et al. Pathophysiological basis of the cardiological benefits of SGLT-2 inhibitors: A narrative review. Cardiovasc Diabetol 2023;22(1):164; doi: 10.1186/s12933-023-01855-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Packer M. SGLT2 inhibitors produce cardiorenal benefits by promoting adaptive cellular reprogramming to induce a state of fasting mimicry: A paradigm shift in understanding their mechanism of action. Diabetes Care 2020;43(3):508–511; doi: 10.2337/dci19-0074 [DOI] [PubMed] [Google Scholar]
- 17. Avogaro A, Fadini GP, Del Prato S. Reinterpreting cardiorenal protection of renal sodium-glucose cotransporter 2 inhibitors via cellular life history programming. Diabetes Care 2020;43(3):501–507; doi: 10.2337/dc19-1410 [DOI] [PubMed] [Google Scholar]
- 18. Griffin M, Rao VS, Ivey-Miranda J, et al. Empagliflozin in heart failure: Diuretic and cardiorenal effects. Circulation 2020;142(11):1028–1039; doi: 10.1161/CIRCULATIONAHA.120.045691 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Pahud de Mortanges A, Salvador D Jr, Laimer M, et al. The role of SGLT2 inhibitors in atherosclerosis: A narrative mini-review. Front Pharmacol 2021;12:751214; doi: 10.3389/fphar.2021.751214 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Mukhopadhyay P, Sanyal D, Chatterjee P, et al. SGLT2 inhibitors: Effect on myocardial infarction and stroke in type 2 diabetes. J Clin Endocrinol Metab 2023;108(8):2134–2140; doi: 10.1210/clinem/dgad113 [DOI] [PubMed] [Google Scholar]
- 21. Nelson AJ, Harrington JL, Kolkailah AA, et al. Sodium-glucose cotransporter-2 inhibitors: Impact on atherosclerosis and atherosclerotic cardiovascular disease events. Heart Fail Clin 2022;18(4):597–607; doi: 10.1016/j.hfc.2022.03.007 [DOI] [PubMed] [Google Scholar]
- 22. Ferrannini E, Solini A, Baldi S, et al. Role of glycosuria in SGLT2 inhibitor-induced cardiorenal protection: A mechanistic analysis of the CREDENCE trial. Diabetes 2024;73(2):250–259; doi: 10.2337/db23-0448 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Sun P, Wang Y, Ding Y, et al. Canagliflozin attenuates lipotoxicity in cardiomyocytes and protects diabetic mouse hearts by inhibiting the mTOR/HIF-1α pathway. iScience 2021;24(6):102521; doi: 10.1016/j.isci.2021.102521 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Verma S, Mudaliar S, Greasley PJ. Potential underlying mechanisms explaining the cardiorenal benefits of sodium-glucose cotransporter 2 inhibitors. Adv Ther 2024;41(1):92–112; doi: 10.1007/s12325-023-02652-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Hwang J, Kim DB, Park HJ. Off-target effects of Sodium-Glucose Cotransporter 2 (SGLT-2) inhibitor in cardiovascular disease. Korean Circ J 2020;50(5):458–460; doi: 10.4070/kcj.2020.0066 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Selvaraj S, Fu Z, Jones P, et al. DEFINE-HF Investigators . Metabolomic profiling of the effects of dapagliflozin in heart failure with reduced ejection fraction: DEFINE-HF. Circulation 2022;146(11):808–818; doi: 10.1161/CIRCULATIONAHA.122.060402 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Santos-Gallego CG, Mayr M, Badimon J. SGLT2 inhibitors in heart failure: Targeted metabolomics and energetic metabolism. Circulation 2022;146(11):819–821; doi: 10.1161/CIRCULATIONAHA.122.060805 [DOI] [PubMed] [Google Scholar]
- 28. Hundertmark MJ, Adler A, Antoniades C, et al. Assessment of cardiac energy metabolism, function, and physiology in patients with heart failure taking empagliflozin: The randomized, controlled EMPA-VISION trial. Circulation 2023;147(22):1654–1669; doi: 10.1161/CIRCULATIONAHA.122.062021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Hyatt HW, Zhang Y, Hood WR, et al. Lactation has persistent effects on a mother’s metabolism and mitochondrial function. Sci Rep 2017;7(1):17118; doi: 10.1038/s41598-017-17418-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Gunderson EP, Lewis CE, Lin Y, et al. Lactation duration and progression to diabetes in women across the childbearing years: The 30-year CARDIA study. JAMA Intern Med 2018;178(3):328–337; doi: 10.1001/jamainternmed.2017.7978 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Tigas S, Sunehag A, Haymond MW. Metabolic adaptation to feeding and fasting during lactation in humans. J Clin Endocrinol Metab 2002;87(1):302–307; doi: 10.1210/jcem.87.1.8178 [DOI] [PubMed] [Google Scholar]
- 32. Neville MC, Sawicki VS, Hay WW Jr, Effects of fasting, elevated plasma glucose and plasma insulin concentrations on milk secretion in women. J Endocrinol 1993;139(1):165–173; doi: 10.1677/joe.0.1390165 [DOI] [PubMed] [Google Scholar]
- 33. Prentice AM, Prentice A. Energy costs of lactation. Annu Rev Nutr 1988;8:63–79; doi: 10.1146/annurev.nu.08.070188.000431 [DOI] [PubMed] [Google Scholar]
- 34. Allen JC, Keller RP, Archer P, et al. Studies in human lactation: Milk composition and daily secretion rates of macronutrients in the first year of lactation. Am J Clin Nutr 1991;54(1):69–80; doi: 10.1093/ajcn/54.1.69 [DOI] [PubMed] [Google Scholar]
- 35. Neville MC, Hay WW Jr, Fennessey P. Physiological significance of the concentration of human milk glucose. Protoplasma 1990;159(2–3):118–128. [Google Scholar]
- 36. Postic C, Leturque A, Printz RL, et al. Development and regulation of glucose transporter and hexokinase expression in rat. Am J Physiol 1994;266(4 Pt 1):E548–E559; doi: 10.1152/ajpendo.1994.266.4.E548 [DOI] [PubMed] [Google Scholar]
- 37. Packer M. Foetal recapitulation of nutrient surplus signalling by O-GlcNAcylation and the failing heart. Eur J Heart Fail 2023;25(8):1199–1212; doi: 10.1002/ejhf.2972 [DOI] [PubMed] [Google Scholar]
- 38. Zhao FQ. Biology of glucose transport in the mammary gland. J Mammary Gland Biol Neoplasia 2014;19(1):3–17; doi: 10.1007/s10911-013-9310-8 [DOI] [PubMed] [Google Scholar]
- 39. Obermeier S, Hüselweh B, Tinel H, et al. Expression of glucose transporters in lactating human mammary gland epithelial cells. Eur J Nutr 2000;39(5):194–200; doi: 10.1007/s003940070011 [DOI] [PubMed] [Google Scholar]
- 40. Shennan DB, Peaker M. Transport of milk constituents by the mammary gland. Physiol Rev 2000;80(3):925–951; doi: 10.1152/physrev.2000.80.3.925 [DOI] [PubMed] [Google Scholar]
- 41. Martin Carli JF, Trahan GD, Jones KL, et al. Single cell RNA sequencing of human milk-derived cells reveals sub-populations of mammary epithelial cells with molecular signatures of progenitor and mature states: A novel, non-invasive framework for investigating human lactation physiology. J Mammary Gland Biol Neoplasia 2020;25(4):367–387; doi: 10.1007/s10911-020-09466-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Lemay DG, Ballard OA, Hughes MA, et al. RNA sequencing of the human milk fat layer transcriptome reveals distinct gene expression profiles at three stages of lactation. PLoS One 2013;8(7):e67531; doi: 10.1371/journal.pone.0067531 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Mohammad MA, Hadsell DL, Haymond MW. Gene regulation of UDP-galactose synthesis and transport: Potential rate-limiting processes in initiation of milk production in humans. Am J Physiol Endocrinol Metab 2012;303(3):E365–E376; doi: 10.1152/ajpendo.00175.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Maningat PD, Sen P, Rijnkels M, et al. Gene expression in the human mammary epithelium during lactation: The milk fat globule transcriptome. Physiol Genomics 2009;37(1):12–22; doi: 10.1152/physiolgenomics.90341.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Kahn BB. Lilly lecture 1995. Glucose transport: Pivotal step in insulin action. Diabetes 1996;45(11):1644–1654; doi: 10.2337/diab.45.11.1644 [DOI] [PubMed] [Google Scholar]
- 46. Klip A, McGraw TE, James DE. Thirty sweet years of GLUT4. J Biol Chem 2019;294(30):11369–11381; doi: 10.1074/jbc.REV119.008351 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Ebeling P, Koistinen HA, Koivisto VA. Insulin-independent glucose transport regulates insulin sensitivity. FEBS Lett 1998;436(3):301–303; doi: 10.1016/s0014-5793(98)01149-1 [DOI] [PubMed] [Google Scholar]
- 48. Burnol AF, Ferre P, Leturque A, et al. Effect of insulin on in vivo glucose utilization in individual tissues of anesthetized lactating rats. Am J Physiol 1987;252(2 Pt 1):E183–E188; doi: 10.1152/ajpendo.1987.252.2.E183 [DOI] [PubMed] [Google Scholar]
- 49. Butte NF, Hopkinson JM, Mehta N, et al. Adjustments in energy expenditure and substrate utilization during late pregnancy and lactation. Am J Clin Nutr 1999;69(2):299–307; doi: 10.1093/ajcn/69.2.299 [DOI] [PubMed] [Google Scholar]
- 50. Appiah D, Lewis CE, Jacobs DR, et al. The association of lactation duration with visceral and pericardial fat volumes in parous women: The CARDIA study. J Clin Endocrinol Metab 2021;106(6):1821–1831; doi: 10.1210/clinem/dgaa980 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Ajmera VH, Terrault NA, VanWagner LB, et al. Longer lactation duration is associated with decreased prevalence of non-alcoholic fatty liver disease in women. J Hepatol 2019;70(1):126–132; doi: 10.1016/j.jhep.2018.09.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Gunderson EP, Hurston SR, Ning X, et al. Study of Women, Infant Feeding and Type 2 Diabetes After GDM Pregnancy Investigators . Lactation and progression to type 2 diabetes mellitus after gestational diabetes mellitus: A prospective cohort study. Ann Intern Med 2015;163(12):889–898; doi: 10.7326/M15-0807 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Gunderson EP, Jacobs DR Jr, Chiang V, et al. Duration of lactation and incidence of the metabolic syndrome in women of reproductive age according to gestational diabetes mellitus status: A 20-year prospective study in CARDIA (Coronary Artery Risk Development in Young Adults). Diabetes 2010;59(2):495–504; doi: 10.2337/db09-1197 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Stuebe AM, Rich-Edwards JW. The reset hypothesis: Lactation and maternal metabolism. Am J Perinatol 2009;26(1):81–88; doi: 10.1055/s-0028-1103034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Poole AT, Vincent KL, Olson GL, et al. Effect of lactation on maternal postpartum cardiac function and adiposity: A murine model. Am J Obstet Gynecol 2014;211(4):424.e1-7; doi: 10.1016/j.ajog.2014.06.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Herrera SR, Vincent KL, Poole A, et al. Long-term effect of lactation on maternal cardiovascular function and adiposity in a murine model. Am J Perinatol 2019;36(5):490–497; doi: 10.1055/s-0038-1669443 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Parikh NI, Gonzalez JM, Anderson CAM, et al. American Heart Association Council on Epidemiology and Prevention; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Cardiovascular and Stroke Nursing; and the Stroke Council . Adverse pregnancy outcomes and cardiovascular disease risk: Unique opportunities for cardiovascular disease prevention in women: A scientific statement from the American Heart Association. Circulation 2021;143(18):e902–e916; doi: 10.1161/CIR.0000000000000961 [DOI] [PubMed] [Google Scholar]
- 58. McClure CK, Schwarz EB, Conroy MB, et al. Breastfeeding and subsequent maternal visceral adiposity. Obesity (Silver Spring) 2011;19(11):2205–2213; doi: 10.1038/oby.2011.185 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Stuebe AM, Rich-Edwards JW, Willett WC, et al. Duration of lactation and incidence of type 2 diabetes. JAMA 2005;294(20):2601–2610; doi: 10.1001/jama.294.20.2601 [DOI] [PubMed] [Google Scholar]
- 60. Fritsche L, Löffler D, Kantartzis K, et al. Effect of breastfeeding duration on coagulation in women with and without history of gestational diabetes mellitus. J Clin Endocrinol Metab 2024;109(10):2571–2578; doi: 10.1210/clinem/dgae172 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Schwarz EB, McClure CK, Tepper PG, et al. Lactation and maternal measures of subclinical cardiovascular disease. Obstet Gynecol 2010;115(1):41–48; doi: 10.1097/AOG.0b013e3181c5512a [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. McClure CK, Catov JM, Ness RB, et al. Lactation and maternal subclinical cardiovascular disease among premenopausal women. Am J Obstet Gynecol 2012;207(1):46.e1-8; doi: 10.1016/j.ajog.2012.04.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Gunderson EP, Quesenberry CP Jr, Ning X, et al. Lactation duration and midlife atherosclerosis. Obstet Gynecol 2015;126(2):381–390; doi: 10.1097/AOG.0000000000000919 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Stuebe AM, Michels KB, Willett WC, et al. Duration of lactation and incidence of myocardial infarction in middle to late adulthood. Am J Obstet Gynecol 2009;200(2):138.e1-8; doi: 10.1016/j.ajog.2008.10.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Tschiderer L, Seekircher L, Kunutsor SK, et al. Breastfeeding is associated with a reduced maternal cardiovascular risk: Systematic review and meta-analysis involving data from 8 studies and 1 192 700 parous women. J Am Heart Assoc 2022;11(2):e022746; doi: 10.1161/JAHA.121.022746 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Ferrannini G, Hach T, Crowe S, et al. Energy balance after sodium-glucose cotransporter 2 inhibition. Diabetes Care 2015;38(9):1730–1735; doi: 10.2337/dc15-0355 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Dewey KG, Heinig MJ, Nommsen LA. Maternal weight-loss patterns during prolonged lactation. Am J Clin Nutr 1993;58(2):162–166; doi: 10.1093/ajcn/58.2.162 [DOI] [PubMed] [Google Scholar]
- 68. Butte NF, King JC. Energy requirements during pregnancy and lactation. Public Health Nutr 2005;8(7A):1010–1027; doi: 10.1079/phn2005793 [DOI] [PubMed] [Google Scholar]
- 69. Pan R, Zhang Y, Wang R, et al. Effect of SGLT-2 inhibitors on body composition in patients with type 2 diabetes mellitus: A meta-analysis of randomized controlled trials. PLoS One 2022;17(12):e0279889; doi: 10.1371/journal.pone.0279889 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Carbone S, O’Keefe JH, Lavie CJ. SGLT2 inhibition, visceral adiposity, weight, and type 2 diabetes mellitus. Obesity (Silver Spring) 2020;28(7):1173; doi: 10.1002/oby.22864 [DOI] [PubMed] [Google Scholar]
- 71. Neeland IJ, de Albuquerque Rocha N, Hughes C, et al. Effects of empagliflozin treatment on glycerol-derived hepatic gluconeogenesis in adults with obesity: A randomized clinical trial. Obesity (Silver Spring) 2020;28(7):1254–1262; doi: 10.1002/oby.22854 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Snyder GG, Holzman C, Sun T, et al. Breastfeeding greater than 6 months is associated with smaller maternal waist circumference up to one decade after delivery. J Womens Health (Larchmt) 2019;28(4):462–472; doi: 10.1089/jwh.2018.7393 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Elliott SA, Pereira LCR, McCargar LJ, et al. ENRICH Study Team . Trajectory and determinants of change in lean soft tissue over the postpartum period. Br J Nutr 2019;121(10):1137–1145; doi: 10.1017/S0007114518002015 [DOI] [PubMed] [Google Scholar]
- 74. Cheong AJY, Teo YN, Teo YH, et al. SGLT inhibitors on weight and body mass: A meta-analysis of 116 randomized-controlled trials. Obesity (Silver Spring) 2022;30(1):117–128; doi: 10.1002/oby.23331 [DOI] [PubMed] [Google Scholar]
- 75. Dardano A, Bianchi C, Garofolo M, et al. The current landscape for diabetes treatment: Preventing diabetes-associated CV risk. Atherosclerosis 2024;394:117560; doi: 10.1016/j.atherosclerosis.2024.117560 [DOI] [PubMed] [Google Scholar]
- 76. Janney CA, Zhang D, Sowers M. Lactation and weight retention. Am J Clin Nutr 1997;66(5):1116–1124; doi: 10.1093/ajcn/66.5.1116 [DOI] [PubMed] [Google Scholar]
- 77. Armenta RF, Kritz-Silverstein D, Wingard D, et al. Association of breastfeeding with postmenopausal visceral adiposity among three racial/ethnic groups. Obesity (Silver Spring) 2015;23(2):475–480; doi: 10.1002/oby.20956 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Neville MC, Keller R, Seacat J, et al. Studies in human lactation: Milk volumes in lactating women during the onset of lactation and full lactation. Am J Clin Nutr 1988;48(6):1375–1386; doi: 10.1093/ajcn/48.6.1375 [DOI] [PubMed] [Google Scholar]
- 79. Butte NF, Garza C, Stuff JE, et al. Effect of maternal diet and body composition on lactational performance. Am J Clin Nutr 1984;39(2):296–306; doi: 10.1093/ajcn/39.2.296 [DOI] [PubMed] [Google Scholar]
- 80. Young BE, Patinkin Z, Palmer C, et al. Human milk insulin is related to maternal plasma insulin and BMI: But other components of human milk do not differ by BMI. Eur J Clin Nutr 2017;71(9):1094–1100; doi: 10.1038/ejcn.2017.75 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Nommsen LA, Lovelady CA, Heinig MJ, et al. Determinants of energy, protein, lipid, and lactose concentrations in human milk during the first 12 mo of lactation: The DARLING study. Am J Clin Nutr 1991;53(2):457–465; doi: 10.1093/ajcn/53.2.457 [DOI] [PubMed] [Google Scholar]
- 82. Kim SY, Yi DY. Components of human breast milk: From macronutrient to microbiome and microRNA. Clin Exp Pediatr 2020;63(8):301–309; doi: 10.3345/cep.2020.00059 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Mohammad MA, Sunehag AL, Chacko SK, et al. Mechanisms to conserve glucose in lactating women during a 42-h fast. Am J Physiol Endocrinol Metab 2009;297(4):E879–E888; doi: 10.1152/ajpendo.00364.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Moon JH, Kim H, Kim H, et al. Lactation improves pancreatic β cell mass and function through serotonin production. Sci Transl Med 2020;12(541):eaay0455; doi: 10.1126/scitranslmed.aay0455 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Ramos-Roman MA, Syed-Abdul MM, Adams-Huet B, et al. Lactation versus formula feeding: Insulin, glucose, and fatty acid metabolism during the postpartum period. Diabetes 2020;69(8):1624–1635; doi: 10.2337/db19-1226 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Neville MC, Casey C, Hay WW Jr, Endocrine regulation of nutrient flux in the lactating woman. Do the mechanisms differ from pregnancy? Adv Exp Med Biol 1994;352:85–98; doi: 10.1007/978-1-4899-2575-6_6 [DOI] [PubMed] [Google Scholar]
- 87. Harreiter J, Vila G, Leitner K, et al. Decreased beta-cell function in breastfeeding obese and non-obese women: A prospective observational study. Clin Nutr 2019;38(6):2790–2798; doi: 10.1016/j.clnu.2018.11.035 [DOI] [PubMed] [Google Scholar]
- 88. McManus RM, Cunningham I, Watson A, et al. Beta-cell function and visceral fat in lactating women with a history of gestational diabetes. Metabolism 2001;50(6):715–719; doi: 10.1053/meta.2001.23304 [DOI] [PubMed] [Google Scholar]
- 89. Bajaj H, Ye C, Hanley AJ, et al. Prior lactation reduces future diabetes risk through sustained postweaning effects on insulin sensitivity. Am J Physiol Endocrinol Metab 2017;312(3):E215–E223; doi: 10.1152/ajpendo.00403.2016 [DOI] [PubMed] [Google Scholar]
- 90. Farahani RA, Egan AM, Welch AA, et al. The effect of glucagon-like peptide 1 receptor blockade on glucagon-induced stimulation of insulin secretion. Diabetes 2023;72(4):449–454; doi: 10.2337/db22-0709 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Pacini G, Ahrén B. Glucagon and GLP-1 exhibit no synergistic enhancement of glucose-stimulated insulin secretion in mice. Peptides 2015;71:66–71; doi: 10.1016/j.peptides.2015.05.010 [DOI] [PubMed] [Google Scholar]
- 92. Noguchi GM, Huising MO. Integrating the inputs that shape pancreatic islet hormone release. Nat Metab 2019;1(12):1189–1201; doi: 10.1038/s42255-019-0148-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Ahrén B, Pacini G. Glucose effectiveness: Lessons from studies on insulin-independent glucose clearance in mice. J Diabetes Investig 2021;12(5):675–685; doi: 10.1111/jdi.13446 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Vahl TP, Paty BW, Fuller BD, et al. Effects of GLP-1-(7-36)NH2, GLP-1-(7-37), and GLP-1-(9-36)NH2 on intravenous glucose tolerance and glucose-induced insulin secretion in healthy humans. J Clin Endocrinol Metab 2003;88(4):1772–1779; doi: 10.1210/jc.2002-021479 [DOI] [PubMed] [Google Scholar]
- 95. Capozzi ME, D’Alessio DA, Campbell JE. The past, present, and future physiology and pharmacology of glucagon. Cell Metab 2022;34(11):1654–1674; doi: 10.1016/j.cmet.2022.10.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Saponaro C, Mühlemann M, Acosta-Montalvo A, et al. Interindividual heterogeneity of SGLT2 expression and function in human pancreatic islets. Diabetes 2020;69(5):902–914; doi: 10.2337/db19-0888 [DOI] [PubMed] [Google Scholar]
- 97. Abdelgani S, Khattab A, Adams J, et al. The impact of increased hepatic glucose production caused by empagliflozin on plasma glucose concentration in individuals with type 2 diabetes and nondiabetic individuals. Diabetes Obes Metab 2024;26(3):1033–1039; doi: 10.1111/dom.15404 [DOI] [PubMed] [Google Scholar]
- 98. Hennart P, Leclercq V, Delogne-Desnoeck J, et al. Plasma glucose, insulin, glucagon and prolactin during long lasting lactation. Horm Metab Res 1984;16(Suppl 1):183–185; doi: 10.1055/s-2007-1014929 [DOI] [PubMed] [Google Scholar]
- 99. Larson-Meyer DE, Schueler J, Kyle E, et al. Do lactation-induced changes in ghrelin, glucagon-like peptide-1, and peptide YY influence appetite and body weight regulation during the first postpartum year? J Obes 2016;2016:7532926; doi: 10.1155/2016/7532926 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. van Gastel N, Carmeliet G. Metabolic regulation of skeletal cell fate and function in physiology and disease. Nat Metab 2021;3(1):11–20; doi: 10.1038/s42255-020-00321-3 [DOI] [PubMed] [Google Scholar]
- 101. Kovacs CS. Maternal mineral and bone metabolism during pregnancy, lactation, and post-weaning recovery. Physiol Rev 2016;96(2):449–547; doi: 10.1152/physrev.00027.2015 [DOI] [PubMed] [Google Scholar]
- 102. Signer RA, Morrison SJ. Mechanisms that regulate stem cell aging and life span. Cell Stem Cell 2013;12(2):152–165; doi: 10.1016/j.stem.2013.01.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Ceccariglia S, Cargnoni A, Silini AR, et al. Autophagy: A potential key contributor to the therapeutic action of mesenchymal stem cells. Autophagy 2020;16(1):28–37; doi: 10.1080/15548627.2019.1630223 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Fernández ÁF, Sebti S, Wei Y, et al. Disruption of the beclin 1-BCL2 autophagy regulatory complex promotes longevity in mice. Nature 2018;558(7708):136–140; doi: 10.1038/s41586-018-0162-7 [DOI] [PMC free article] [PubMed] [Google Scholar]

