Potential role of β‑carotene‑modulated autophagy in puerperal breast inflammation (Review)

  • Authors:
    • Stella Tinia Hasianna
    • Julia Windi Gunadi
    • Enny Rohmawaty
    • Ronny Lesmana
  • View Affiliations

  • Published online on: July 22, 2022     https://doi.org/10.3892/br.2022.1558
  • Article Number: 75
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Puerperal breast inflammation is common in the first 6‑8 weeks postpartum, and without proper management, may lead to a decrease in breastmilk production and early cessation of breastfeeding. Recent studies showed that carotenoids decrease the severity of puerperal breast inflammation. This article summarizes the significant findings on β‑carotene with a potential role as an autophagy modulator in puerperal breast inflammation. Puerperal milk stasis causes an increase in inflammatory cytokines and inflammatory cells, leading to reactive oxygen species (ROS) activation that causes oxidative damage to mammary glands and affects breast milk secretion. β‑carotene has an anti‑inflammatory effect related to its ROS‑scavenging activity and modulates autophagy, thus stimulating the removal of damaged cellular structures and supporting milk gland survival. β‑carotene modulates autophagy through phosphorylation of NF‑κB, JNK, p38, Akt, and Nrf2, affects the ratio of Microtubule‑associated protein 1A/1B‑light chain 3 (LC3)‑II/LC3‑I, and has a role in the regulation of the JAK2/STAT3, PI2K/Akt/mTOR and AMPK pathways. Although the in vitro and in vivo studies showed promising results, further studies on humans are required to better conclude the potential role of β‑carotene in managing puerperal breast inflammation.

1. Introduction

In exclusive breastfeeding (EBF), breast milk becomes the only source of nutrition for infants without any other liquids or solids (1). EBF is beneficial for the baby as well as the mother (2-4), and the World Health Organization (WHO) and United Nations Children's Fund (UNICEF) recommend EBF for the first 6 months of an infant's life and should then be continued for 2 years or more (5). However, in 2020, only 44% of babies under 6 months worldwide received exclusive breastfeeding (2). Many factors influence the cessation of breastfeeding, including insufficient milk supply (6-8), self-weaning (9), primiparity (10), mother/infant separation (6), inconvenience/fatigue due to breastfeeding (11), and inflammatory breast diseases such as mastitis (12,13).

Mastitis is inflammation of the breast, with or without infection (14). Worldwide, -20% of women suffer from mastitis whilst breastfeeding (15). The majority of cases occur in the first 6-8 weeks postpartum, and approximately one-third of women experience a recurrent episode (16,17). Up to 3% of cases of mastitis develop into a breast abscess (18). Puerperal breast inflammation starts with insufficient breast emptying (milk stasis) (12). This causes an increase in intraductal pressure and opens milk duct epithelial cells' intercellular junctions. Breast milk further moves into the connective tissue and generates a sterile inflammatory environment, from which a secondary bacterial infection may follow. Without proper treatment, this can progress to a mammary abscess that requires surgical treatment (12). Several studies indicate that puerperal breast inflammation is correlated with the cessation of breastfeeding (12,16).

Milk stasis increases the levels of inflammatory cytokines (TNF-α, IL-1b, IL-6, IL-8), neutrophils, macrophages, lymphocytes, eosinophils, and various epithelial cells of mammary tissue (19). Cytokines further activate reactive oxygen species (ROS), which play a complex role in the inflammatory process (20). The excessive buildup of ROS disrupts cellular homeostasis, causes oxidative stress and mitochondrial dysfunction, and induces autophagy (21). The oxidative damage in breast inflammation can cause mammary gland cell death and affects breast milk secretions (15).

Antioxidant supplementation decreases the incidence, duration, and severity of mastitis (22,23). Antioxidants scavenge free radicals and inhibit the activity of oxidizing enzymes, thus reducing the damage caused by free radicals (23). Some herbal treatments with high antioxidant and anti-inflammatory activities are widely used in mastitis treatment, such as Moringa leaves (24), binahong leaves (Anredera cordifolia) (25), cherry leaf decoction (Muntingia calabura L.) (26), Red Algae Eucheuma Spinosum (27), Macroalgae Extract (28), garlic, manjakani fruit, betel leaf, white turmeric, and eucalyptus leaves (29). Carotenoids are another type of active substance found in plants with high antioxidant contents and anti-inflammatory effects, but have not been as widely explored as treatment or prevention options for mastitis. Previous studies showed that increasing serum retinol concentrations in dairy cows was associated with a decreased risk of clinical mastitis, and low concentrations of vitamin A and β-carotene increased the severity of mastitis (30,31). Carotenoids have a significant impact on maintaining the health of epithelial tissue and the stability of mucosal surface integrity (32).

Carotenoids are yellow-orange pigments that are abundantly present in several plants, marine invertebrates, and microorganisms. There are ~50 carotenoids consumed in a standard human diet, but only 20 have been identified in the human plasma, with β-carotene, α-carotene, lycopene, and cryptoxanthin as the most common (33). Carotenoids have potent antioxidant, anti-inflammatory, and immunomodulatory abilities. (34) β-carotene is the most common carotenoid, which is also known as pro-vitamin A, and is one of the most abundant carotenoids in the human blood with several health-promoting properties (35,36). The anti-inflammatory effects of β-carotene have been demonstrated in multiple systems (37,38).

Previous studies showed that β-carotene and vitamin A reduce the risk of puerperal breast inflammation (39). Low concentrations of plasma vitamin A (<80 µg/100 ml) and β-carotene (<200 µg/100 ml) correlated with the severity of mastitis (23,30). Animal studies in cows showed that mastitic cows, as determined using the California Mastitis Test score, had significantly lower plasma vitamin A and β-carotene concentrations than healthy cows (32). To reduce the risk of mastitis, a dry and lactating cow's diet should contain 110 IU/kg bwt/day of vitamin A (22). Plasma concentrations of β-carotene >3 mg/l promoted udder health in dairy cattle (40). Low concentrations of plasma vitamin A (<0.8 µg/ml) and β-carotene (<2 µg/ml) were linked with the severity of mastitis. However, unfortunately, there is minimal data regarding the optimal dose of β-carotene for postpartum women in correlation to treatment or prevention of puerperal breast inflammation.

As an antioxidant, β-carotene suppresses intracellular ROS production and reduces antioxidative enzyme activity (36,41,42). Β-carotene supplementation exerts a stabilizing effect on polymorphonuclear cells such as neutrophils, eosinophils, and basophils, and optimizes lymphocyte function (23). The inflammatory response in mastitis and the role of β-carotene as an antioxidant is summarized in Fig. 1. However, there is limited study on the role of β-carotene in correlation with autophagy modulation and puerperal breast inflammation. The present review summarizes the major findings on β-carotene as a potential autophagy modulator in puerperal breast inflammation.

2. Autophagy in puerperal breast inflammation

Breast fullness or breast engorgement is a part of lactogenesis in the postpartum period. This fullness manifests ~36 h postpartum and occurs 3-7 days postpartum, with primiparous mothers most commonly affected (43). In breast engorgement, the breast undergoes increased vascularity, congestion, milk accumulation, and oedema (43,44).

In milk stasis, the accumulation of breast milk disrupts vascularisation and causes tissue hypoxia (19). Tissue hypoxia induces the transcription of Bcl2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3) and induction of NIX following HIF1 activation (21). This in turn competes with beclin-1 for the binding of Bcl-2, and the release of beclin-1 is triggered, followed by autophagy (45). PERK, an ER stress sensor, is stimulated and, later in the process, stimulates the expression of Microtubule-associated protein 1A/1B-light chain 3 (LC3) and autophagy gene (ATG)5, both of which are ATGs (46). Oxidative stress triggers FOXO3 and then stimulates the transcription of LC3, BNIP, and NRF2, which induces transcription of p62(21). The process mentioned above positively modulates autophagy. p53 has a role in activating DNA damage-regulated autophagy modulator and sestrins, two autophagy-related genes that positively regulate autophagy (47). TP53-Induced Glycolysis and Apoptosis Regulator negatively regulates autophagy, but while it is a p53 target, it possesses a p53-independent function in autophagy (21). AMPK is also p53-independent, and is activated by sestrins to inhibit mTOR activity and thus induce autophagy (48). ROS constrains ATG4 protease activity and promotes autophagosome formation (21,49).

Milk stasis also results in high oxygen and energy demands and increases ROS production as a result (12). ROS can assist the healing and tissue repair process in moderate concentrations, but in excess, ROS disrupts redox homeostasis and this results in oxidative stress and damage to cell organelles (50). The increase in free radical levels in puerperal breast inflammation is triggered by the increase of neutrophils, macrophages, lymphocytes, eosinophils, and mammary tissue epithelial cells, TNF-α, IL-1b, IL-6, IL-8, and nitric oxide (NO) (51). ROS accumulation triggers endothelial dysfunction and tissue injury in mammary gland tissue, opens inter-endothelial junctions, and stimulates inflammatory cell migration across the endothelial barrier. These cells support the clearance of pathogens, but excessive amounts may also cause tissue injury (52).

ROS is essential in promoting cell autophagy as a critical signalling molecule (53). Autophagy facilitates the degradation of intracellular proteins and organelles via lysosomes. ROS induces autophagy, and autophagy reduces oxidative damage (49). During inflammation, the NRF2 transcription factor enhances p62 expression, and p62, in turn, creates a positive feedback loop for NRF2(49).

Autophagy is involved in inflammatory reactions and barrier repair by degrading cell components via the lysosomal pathway (54). Autophagosomes are formed during autophagy to encapsulate protein aggregates and/or damaged organelles. Hydrolytic enzymes degrade the damaged organelles after autophagosome fusion with a lysosome (55). Autophagy is stimulated by any stressor that causes cellular stress, including hypoxia (56), nutrient deficiency (57), chemical exposure (56), excessive ROS (58), or intracellular pathogens (59). Autophagy is a cellular recycling process that involves potential cell death outcomes and functions to protect cells from apoptosis (60).

Four sub-groups of ATGs regulate autophagy; namely Atg1/unc-51-like kinase (ULK) complex, two ubiquitin-like proteins [Atg12 and Atg8/microtubule-associated protein light chain 3 (LC3)] conjugation systems, the class III phosphatidylinositol 3-kinase (PI3K)/vacuolar protein sorting 34 (Vps34) complex I, and two transmembrane proteins, Atg9/mAtg9 and VMP1(61). ROS induces autophagy through two methods, directly and indirectly. The direct regulation is established through modulation of the involved proteins, such as Atg4, Atg5, and Beclin. Indirect regulation is achieved through autophagy-related signaling pathways, such as the mTOR, mitogen-activated protein kinase (MAPK), p38, ERK, and JNK (62). ROS inhibits mTOR activity and activates AMPK, and initiates autophagy through the increase of Vps34 complex activity (50). Activation of the p53 pathway causes JNK and Sestrin2 activation. JNK and Sestrin2 bind to TSC1/TSC2 and induce autophagy (62). ROS also promotes LC3-II translocation through the inhibition of ATG4, which converts LC3-II into LC3-I (63).

Excessive ROS production triggers cell death extrinsically and intrinsically (52). The extrinsic pathway is mediated via four major cell death receptors; TNF receptor 1, TNF-related apoptosis-inducing ligand receptor (TRAIL-R)1, TRAIL-R2, and the Fas receptor (19). TNFα, Fas ligand, and TRAIL bind to these receptors. TNFα is secreted by activated macrophages and is involved in the increase in inflammatory cytokines (50).

The increase in mitochondrial outer membrane permeability (MOMP) triggers intrinsic pathways of cell death (52). MOMP induces the release of Cytochrome-c (Cyt-c), Smac/Diablo, apoptosis-inducing factor (AIF), and endonuclease G, which play a role in cell death in either a caspase-dependent or -independent manner (64). This increase in MOMP is stimulated by an excessive entry of calcium and a high level of oxidative stress, which causes mitochondrial membrane depolarization (52). An apoptosome is formed as Cyt-c attaches to apoptosis activation factor-1 and this recruits the initiator pro-caspase 9. Caspase-9 functions to trigger caspase-3. (58) Smac/Diablo also plays a role in activating effector caspases by eliminating the blockage of the inhibitor of apoptosis proteins (50). After AIF is activated, DNA condensation and cleavage induction occur (52). The primary determinant of cell death is the mitochondrial release of Cyt-c. BH3 interacting-domain death agonist, a pro-apoptotic member of the Bcl-2 family, cleaves caspase-8 and increases MOMP, followed by oligomerization of pro-apoptotic proteins Bax/Bak which induce Cyt-c release (52).

The release of Cyt-c is triggered by oxidative stress, facilitates electron transport chain uncoupling, and increases mitochondrial ROS production/levels. Cardiolipin (CL) is an anionic phospholipid that aids Cyt-c anchoring to the inner mitochondrial membrane. The affinity binding with Cyt-c is reduced in oxidative stress due to CL oxidation. CL-bound mitochondrial Cyt-c catalyses CL peroxidation, and this phenomenon can occur at a lower H2O2 concentration when bound to CL-containing membranes. Thus, oxidative stress stimulates cell death through the peroxidase activity of Cyt-c and CL peroxidation (52).

3. The role of β-carotene in the modulation of autophagy in puerperal breast inflammation

Mastitis is related to an inadequate intake of vitamin E, selenium, β-carotene, and vitamin A (65). Antioxidants decrease the duration, incidence, and severity of mastitis (23). Β-carotene and vitamin A can protect mammary tissues and milk from the destructive effects of free radicals. Food containing vitamin A and β-carotene contributed to a reduced incidence of mastitis during the early dry period in cows (23).

Carotene exhibits a range of important functions, and the numerous benefits of carotenoids are related to their antioxidant and anti-inflammatory effects. Vitamin A and β-carotene trigger the generation of immune cells and prevent the initiation of the fatty acid peroxidation chain reaction (39). Antioxidants directly scavenge free radicals and inhibit the activity of oxidizing enzymes, thus protecting the body from free radicals (23). β-carotene quenches singlet oxygen and neutralizes lipid peroxyl radicals. It has been previously shown that 800 mg/kg body weight of algae Spirulina fusiformis, containing β-carotene and SOD, reduced the levels of almost all oxidative stress biomarkers measured in the study. Serum lipid hydroperoxide levels were reduced by 45%, and leukocyte MDA levels were reduced by 50% (66). β-carotene has a long system of conjugated double bonds with π-electrons delocalized over the length of the polyene chain, providing it with an effective ability to scavenge ROS (67). Studies have shown that β-carotene significantly inhibited intracellular ROS production (30,32,42).

In a study investigating the effects of β-carotene in corneal endothelial cells, a film scaffold incorporated with an appropriate amount of β-carotene showed enhanced initial cell adhesion, proliferation, proper cell morphology, and gene expression compared with a pristine silk fibroin (SF) scaffold. β-carotene in the SF film scaffold enhanced the function of the ATPase pump of corneal endothelial cells (68). In infected tissues, β-carotene decreases the expression of inflammatory mediators, MAPKs, and redox-sensitive transcription factors (69).

β-carotene is widely available in common foodstuffs with broad affordability for the general community. Using β-carotene for postpartum mothers can reduce the occurrence of severe mastitis and maternal and infant morbidity and mortality (32). Although β-carotene and vitamin A serve essential roles in immunity, the latest recommendation from WHO states that vitamin A supplementation for postpartum mothers is not recommended as a public health intervention to prevent maternal and infant morbidity and mortality. Postpartum mothers must obtain a balanced healthy nutritional intake to obtain an adequate amount of various nutrients needed (70,71). Dietary dosage of carotenoids may promote health, but supplementation with high doses may be associated with adverse effects, especially in smokers or subjects exposed to environmental pollutants (72).

β-carotene is abundantly present in green leafy vegetables, carrots, pumpkins, papaya and red palm oil, milk, liver, and fish oil (71). It has been shown that consuming fruit and vegetable sources of β-carotene increases the vitamin A status of lactating women (73). Vitamin A is recommended at 1,300 mcg retinol activity equivalent (RAE) daily. One mcg RAE is equivalent to 2 mcg supplemental β-carotene or 12 mcg dietary β-carotene (74). However, several factors affect the bioavailability and equivalency of vitamin A, such as food processing techniques and the specific dietary intake of an individual (73).

β-carotene plays an essential role in modulating autophagy. In LPS-induced rat intestinal tissue, β-carotene treatment significantly reduced the LPS-mediated phosphorylation of JAK2/STAT3 and NF-κB (59), and significantly suppressed the LPS-induced phosphorylation of JNK and p38(41) The levels of p-Akt are decreased following LPS treatment, and β-carotene significantly upregulated the phosphorylation of Akt. (41) It also significantly suppressed the ratio of LC3-II/LC3-I, which was upregulated by LPS (41). LC3 consists of LC3-I and LC3-II. LC3-II is positively correlated with the degree of autophagy, and is used as an autophagy marker. LC3-I is present in the cytoplasm where a small segment of the polypeptide is cleaved and is converted into LC3-II (75). β-carotene modulates LPS-induced autophagy and inhibits the activity of the related inflammation pathways (41).

However, another study showed that oral β-carotene supplementation daily for 14 days in male ddY mice (8 wk old) did not alter the ratio of LC3-II to LC3-I in the β-carotene group (76). In H9C2 cell lines, low-dose β-carotene induces autophagy, as shown by a decrease in LC3II and p62 levels. NF-κB protein levels were lowered, and Nrf2 was triggered, but no significant alteration of Nrf1 was observed. A low dose of βcarotene stimulates cell viability by suppressing the apoptotic signals carried by caspase 3 and 9. Furthermore, a low dose of β-carotene increases cell viability through autophagy stimulation, inhibition of pro-inflammatory factors, and suppression of apoptosis (36).

Treatment of SAOs and Caco-2, cell lines derived from a human osteosarcoma and colon adenocarcinoma, respectively, using 200-400 µg/ml carotene extract nanoemulsion (CEN) triggered autophagy, based on the increase of autophagosome formation, increased expression of LC3-II, modulation of autophagic flux, increased phosphorylation, and an increase in the levels of the active form of AMPK kinase pAMPKThr172, which activates AMPK and induces autophagy (33). β-carotene is the major carotenoid in CEN (33). However, treatment with the same dose of β-carotene alone did not show the same effect as when combined in the CEN, indicating a dose-dependent effect or a synergism of different components in CEN (33).

The PI3K/Akt/mTOR signaling pathway is important in cell survival and autophagy (77). Inhibition of this signaling pathway induces autophagy. Consistent with previous studies, β-carotene treatment was also shown to decrease Advanced Glycation End products (AGE)-induced elevation of the LC3II/LC3I ratio and the number of LC3-labeled puncta. The protective effect of β-carotene in AGE-induced H9c2 cells was achieved through the activation of the PI3K/Akt/mTOR signaling pathway (42). Fig. 2 summarizes the pathways affected by β-carotene as an autophagy modulator.

β-carotene shows promising potential as an autophagy modulator in increasing mammary gland cell survival and maintaining breastmilk production. Further research is required to understand the role of β-carotene in puerperal breast inflammation management.

4. Conclusion

Puerperal breast inflammation can cause result in cessation of breastfeeding due to mammary gland damage and a decrease in milk production. Excessive ROS levels in puerperal breast inflammation results in oxidative damage and induces improper autophagy. β-carotene has ROS-scavenging activity and the ability to modulate autophagy through the suppression of JAK2/STAT3, NF-κB, JNK, and p38 phosphorylation, suppressing the ratio of LC3-II/LC3-I and upregulating the phosphorylation of Akt. Further studies are required to conclude the full potential of β-carotene in puerperal breast inflammation.

Acknowledgements

Not applicable.

Funding

Funding: This study was supported by a WCR grant from the Ministry of Education and Culture (grant no. 1207/UN6.3.1/PT.00/2021) and an internal grant from the Universitas Kristen Maranatha (grant no. 034/SK/ADD/UKM/VI/2021).

Availability of data and materials

Not applicable.

Authors' contributions

STH, RL, JWG, and ER performed the literature search and assisted in drafting and revising the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Silva FV, Dias F, Costa G and Campos MDG: Chamomile reveals to be a potent galactogogue: The unexpected effect. J Matern Neonatal Med. 31:116–118. 2018.PubMed/NCBI View Article : Google Scholar

2 

World Health Organization (WHO) and United Nations Children's Fund (UNICEF): Indicators for Assessing Infant and Young Child Feeding Practices. Vol. WHA55 A55/. WHO, Geneva, p122, 2021. http://apps.who.int/iris/bitstream/handle/10665/44306/9789241599290_eng.pdf?sequence=1%0Ahttp://whqlibdoc.who.int/publications/2008/9789241596664_eng.pdf%5Cnhttp://www.unicef.org/programme/breastfeeding/innocenti.htm%5Cnhttp://innocenti15.net/declaration.

3 

Mosca F and Giannì ML: Human milk: Composition and health benefits. Pediatr Med Chir. 39(155)2017.PubMed/NCBI View Article : Google Scholar

4 

Henrick BM, Nag K, Yao XD, Drannik AG, Aldrovandi GM and Rosenthal KL: Milk matters: Soluble toll-like receptor 2 (sTLR2) in breast milk significantly inhibits HIV-1 infection and inflammation. PLoS One. 7(e40138)2012.PubMed/NCBI View Article : Google Scholar

5 

World Health Organization and United Nations Children's Fund (UNICEF): Baby-friendly hospital initiative: revised, updated and expanded for integrated care. WHO, Geneva, 2009. https://apps.who.int/iris/handle/10665/43593.

6 

Chang PC, Li SF, Yang HY, Wang LC, Weng CY, Chen KF, Chen W and Fan SY: Factors associated with cessation of exclusive breastfeeding at 1 and 2 months postpartum in Taiwan. Int Breastfeed J. 14(18)2019.PubMed/NCBI View Article : Google Scholar

7 

Grzeskowiak LE, Wlodek ME and Geddes DT: What evidence do we have for pharmaceutical galactagogues in the treatment of lactation insufficiency?-a narrative review. Nutrients. 11(974)2019.PubMed/NCBI View Article : Google Scholar

8 

Lou Z, Zeng G, Huang L, Wang Y, Zhou L and Kavanagh KF: Maternal reported indicators and causes of insufficient milk supply. J Hum Lact. 30:466–473; quiz 511-2. 2014.PubMed/NCBI View Article : Google Scholar

9 

Brown CRL, Dodds L, Legge A, Bryanton J and Semenic S: Factors influencing the reasons why mothers stop breastfeeding. Can J Public Health. 105:e179–e185. 2014.PubMed/NCBI View Article : Google Scholar

10 

Grzeskowiak LE, Dalton JA and Fielder AL: Factors associated with domperidone use as a galactogogue at an australian tertiary teaching hospital. J Hum Lact. 31:249–253. 2015.PubMed/NCBI View Article : Google Scholar

11 

Riordan J, Bibb D, Miller M and Rawlins T: Predicting breastfeeding duration using the LATCH breastfeeding assessment tool. J Hum Lact. 17:20–23. 2001.PubMed/NCBI View Article : Google Scholar

12 

Wöckel A, Abou-Dakn M, Beggel A and Arck P: Inflammatory breast diseases during lactation: Health effects on the newborn-A literature review. Mediators Inflamm. 2008(298760)2008.PubMed/NCBI View Article : Google Scholar

13 

Schwartz K, D'Arcy HJ, Gillespie B, Bobo J, Longeway M and Foxman B: Factors associated with weaning in the first 3 months postpartum. J Fam Pract. 51:439–444. 2002.PubMed/NCBI

14 

Bond DM, Morris JM and Nassar N: Study protocol: Evaluation of the probiotic Lactobacillus Fermentum CECT5716 for the prevention of mastitis in breastfeeding women: A randomised controlled trial. BMC Pregnancy Childbirth. 17(148)2017.PubMed/NCBI View Article : Google Scholar

15 

Pevzner M and Dahan A: Mastitis while breastfeeding: Prevention, the importance of proper treatment, and potential complications. J Clin Med. 9(2328)2020.PubMed/NCBI View Article : Google Scholar

16 

Scott JA, Robertson M, Fitzpatrick J, Knight C and Mulholland S: Occurrence of lactational mastitis and medical management: A prospective cohort study in Glasgow. Int Breastfeed J. 3(21)2008.PubMed/NCBI View Article : Google Scholar

17 

Cooklin AR, Amir LH, Nguyen CD, Buck ML, Cullinane M, Fisher JRW and Donath SM: CASTLE Study Team. Physical health, breastfeeding problems and maternal mood in the early postpartum: A prospective cohort study. Arch Womens Ment Health. 21:365–374. 2018.PubMed/NCBI View Article : Google Scholar

18 

Royster E: Milk quality and mastitis. 1–33. 2016.

19 

Sordillo LM: Factors affecting mammary gland immunity and mastitis susceptibility. Livest Prod Sci. 98:89–99. 2005.

20 

Li C, Solomons NW, Scott ME and Koski KG: Subclinical mastitis (SCM) and proinflammatory cytokines are associated with mineral and trace element concentrations in human breast milk. J Trace Elem Med Biol. 46:55–61. 2018.PubMed/NCBI View Article : Google Scholar

21 

Li L, Tan J, Miao Y, Lei P and Zhang Q: ROS and Autophagy: Interactions and molecular regulatory mechanisms. Cell Mol Neurobiol. 35:615–621. 2015.PubMed/NCBI View Article : Google Scholar

22 

Yang FL and Li XS: Role of antioxidant vitamins and trace elements in mastitis in dairy cows. J Adv Vet Anim Res. 2:1–9. 2015.

23 

Ellah MR: Role of Free Radicals and Antioxidants in Mastitis. J Adv Vet Res. 3:1–7. 2013.

24 

Kurnianto D: Inhibitory power of moringa leaf juice (Moringa oleifera) against Staphylococcus aureus and Escherichia coli growth as the etiology of mastitis in cattle (unpublished PhD thesis). Universitas Brawijaya, 2015.

25 

Wijayanti D and Ardigurnita F: The development of binahong leaf as an antiseptic to overcome mastitis in dairy cows in the Giri Mukti herd, Tasikmalaya. J-Dinamika Journal of Public Health. 4:148–152. 2019.https://publikasi.polije.ac.id/index.php/j-dinamika/article/view/1087.

26 

Kurniawan I, Sarwiyono S and Surjowardojo P: The effect of teat dipping using cherry leaf decoction (Muntingia calabura L.) on the incidence of mastitis. Indonesian Journal of Animal Science. 23:27–31. 2010.

27 

Xiao H, Zhao J, Fang C, Cao Q, Xing M, Li X, Hou J, Ji A and Song S: Advances in studies on the pharmacological activities of fucoxanthin. Mar Drugs. 18(634)2020.PubMed/NCBI View Article : Google Scholar

28 

Putri SU: Effect of Macroalgae extract on bacteria Staphylococcus aureus and methicillin resistant Staphylococcus aureus (unpublished PhD thesis). Alauddin Islamic State University, 2016.

29 

Triandini IGAAH, Ruqqayah S and Astuti NLB: In vitro research on plants as natural antibiotics for breast inflammation (Mastitis). Sangkareang Mataram Scientific Journal. 4(3):14–17. 2018.https://sangkareang.org/index.php/SANGKAREANG/article/view/122.

30 

Leblanc SJ, Herdt TH, Seymour WM, Duffield TF and Leslie KE: Peripartum serum vitamin E, retinol, and beta-carotene in dairy cattle and their associations with disease. J Dairy Sci. 87:609–619. 2004.PubMed/NCBI View Article : Google Scholar

31 

Chew BP, Hollen LL, Hillers JK and Herlugson ML: Relationship between vitamin A and β-carotene in blood plasma and milk and mastitis in holsteins. J Dairy Sci. 65:2111–2118. 1982.PubMed/NCBI View Article : Google Scholar

32 

Heinrichs AJ, Costello SS and Jones CM: Control of heifer mastitis by nutrition. Vet Microbiol. 134:172–176. 2009.PubMed/NCBI View Article : Google Scholar

33 

Russo M, Moccia S, Bilotto S, Spagnuolo C, Durante M, Lenucci MS, Mita G, Volpe MG, Aquino RP and Russo GL: A carotenoid extract from a Southern Italian cultivar of pumpkin triggers nonprotective autophagy in malignant cells. Oxid Med Cell Longev. 2017(7468538)2017.PubMed/NCBI View Article : Google Scholar

34 

Zielińska MA, Wesołowska A, Pawlus B and Hamułka J: Health effects of carotenoids during pregnancy and lactation. Nutrients. 9(838)2017.PubMed/NCBI View Article : Google Scholar

35 

Crackers NR and Per V: USDA National Nutrient Database for Standard Reference. Release 1 (Omega 3). 3:3–7. 2019.https://ods.od.nih.gov/pubs/usdandb/VitA-betaCarotene-Content.pdf.

36 

Lesmana R, Felia Yusuf I, Goenawan H, Achadiyani A, Khairani AF, Nur Fatimah S and Supratman U: Low Dose of β-carotene regulates inflammation, reduces caspase signaling, and correlates with autophagy activation in cardiomyoblast cell lines. Med Sci Monit Basic Res. 26(e928648)2020.PubMed/NCBI View Article : Google Scholar

37 

Cheng J, Balbuena E, Miller B and Eroglu A: The role of β-carotene in colonic inflammation and intestinal barrier integrity. Front Nutr. 8(723480)2021.PubMed/NCBI View Article : Google Scholar

38 

Palozza P: Can beta-carotene regulate cell growth by a redox mechanism? An answer from cultured cells. Biochim Biophys Acta. 1740:215–221. 2005.PubMed/NCBI View Article : Google Scholar

39 

Janik IA: The detection and prediction of mastitis in dairy cows by particle analysis (unpublished PhD thesis). Coventry University, 2013.

40 

O'Rourke D: Nutrition and udder health in dairy cows: A review. Ir Vet J. 62 (Suppl 4):S15–S20. 2009.PubMed/NCBI View Article : Google Scholar

41 

Yang Y, Li R, Hui J, Li L and Zheng X: β-Carotene attenuates LPS-induced rat intestinal inflammation via modulating autophagy and regulating the JAK2/STAT3 and JNK/p38 MAPK signaling pathways. J Food Biochem. 45(e13544)2021.PubMed/NCBI View Article : Google Scholar

42 

Zhao G, Zhang X, Wang H and Chen Z: Beta carotene protects H9c2 cardiomyocytes from advanced glycation end product-induced endoplasmic reticulum stress, apoptosis, and autophagy via the PI3K/Akt/mTOR signaling pathway. Ann Transl Med. 8(647)2020.PubMed/NCBI View Article : Google Scholar

43 

Mass S: Breast pain: Engorgement, nipple pain and mastitis. Clin Obstet Gynecol. 47:676–682. 2004.PubMed/NCBI View Article : Google Scholar

44 

Guo W, Liu B, Yin Y, Kan X, Gong Q, Li Y, Cao Y, Wang J, Xu D, Ma H, et al: Licochalcone A protects the blood milk barrier integrity and relieves the inflammatory response in LPS-Indued mastitis. Front Immunol. 10(287)2019.PubMed/NCBI View Article : Google Scholar

45 

Zhu Y, Cheng J, Min Z, Yin T, Zhang R, Zhang W, Hu L, Cui Z, Gao C, Xu S, et al: Effects of fucoxanthin on autophagy and apoptosis in SGC-7901cells and the mechanism. J Cell Biochem. 119:7274–7284. 2018.PubMed/NCBI View Article : Google Scholar

46 

Zeng M, Sang W, Chen S, Chen R, Zhang H, Xue F, Li Z, Liu Y, Gong Y, Zhang H and Kong X: 4-PBA inhibits LPS-induced inflammation through regulating ER stress and autophagy in acute lung injury models. Toxicol Lett. 271:26–37. 2017.PubMed/NCBI View Article : Google Scholar

47 

Wang X, Su F, Yu X, Geng N, Li L, Wang R, Zhang M, Liu J, Liu Y and Han B: RNA-Seq whole transcriptome analysis of bovine mammary epithelial cells in response to intracellular staphylococcus aureus. Front Vet Sci. 7(642)2020.PubMed/NCBI View Article : Google Scholar

48 

Hou LL, Gao C, Chen L, Hu GQ and Xie SQ: Essential role of autophagy in fucoxanthin-induced cytotoxicity to human epithelial cervical cancer HeLa cells. Acta Pharmacol Sin. 34:1403–1410. 2013.PubMed/NCBI View Article : Google Scholar

49 

Scherz-Shouval R and Elazar Z: Regulation of autophagy by ROS: Physiology and pathology. Trends Biochem Sci. 36:30–38. 2011.PubMed/NCBI View Article : Google Scholar

50 

He L, He T, Farrar S, Ji L, Liu T and Ma X: Antioxidants maintain cellular redox homeostasis by elimination of reactive oxygen species. Cell Physiol Biochem. 44:532–553. 2017.PubMed/NCBI View Article : Google Scholar

51 

Atakisi O, Oral H, Atakisi E, Merhan O, Metin Pancarci S, Ozcan A, Marasli S, Polat B, Colak A and Kaya S: Subclinical mastitis causes alterations in nitric oxide, total oxidant and antioxidant capacity in cow milk. Res Vet Sci. 89:10–13. 2010.PubMed/NCBI View Article : Google Scholar

52 

Mittal M, Siddiqui MR, Tran K, Reddy SP and Malik AB: Reactive oxygen species in inflammation and tissue injury. Antioxidants Redox Signal. 20:1126–1167. 2014.PubMed/NCBI View Article : Google Scholar

53 

Cao Y, Wang J, Tian H and Fu GH: Mitochondrial ROS accumulation inhibiting JAK2/STAT3 pathway is a critical modulator of CYT997-induced autophagy and apoptosis in gastric cancer. J Exp Clin Cancer Res. 39(119)2020.PubMed/NCBI View Article : Google Scholar

54 

Guo W, Li W, Su Y, Liu S, Kan X, Ran X, Cao Y, Fu S and Liu J: GPR109A alleviate mastitis and enhances the blood milk barrier by activating AMPK/Nrf2 and autophagy. Int J Biol Sci. 17:4271–4284. 2021.PubMed/NCBI View Article : Google Scholar

55 

Qian M, Fang X and Wang X: Autophagy and inflammation. Clin Transl Med. 6(24)2017.PubMed/NCBI View Article : Google Scholar

56 

Sun Y, Huang YH, Huang FY, Mei WL, Liu Q, Wang CC, Lin YY, Huang C, Li YN, Dai HF and Tan GH: 3'-epi-12β-hydroxyfroside, a new cardenolide, induces cytoprotective autophagy via blocking the Hsp90/Akt/mTOR axis in lung cancer cells. Theranostics. 8:2044–2060. 2018.PubMed/NCBI View Article : Google Scholar

57 

Jung CH, Ro SH, Cao J, Otto NM and Kim DH: MTOR regulation of autophagy. FEBS Lett. 584:1287–1295. 2010.PubMed/NCBI View Article : Google Scholar

58 

Chang KC, Liu PF, Chang CH, Lin YC, Chen YJ and Shu CW: The interplay of autophagy and oxidative stress in the pathogenesis and therapy of retinal degenerative diseases. Cell Biosci. 12(1)2022.PubMed/NCBI View Article : Google Scholar

59 

Hadad N and Levy R: The synergistic anti-inflammatory effects of lycopene, lutein, β-carotene, and carnosic acid combinations via redox-based inhibition of NF-κB signaling. Free Radic Biol Med. 53:1381–1391. 2012.PubMed/NCBI View Article : Google Scholar

60 

Lee H, Lim JW and Kim H: Effect of Astaxanthin on Activation of Autophagy and Inhibition of Apoptosis in Helicobacter pylori-Infected Gastric Epithelial Cell Line AGS. Nutrients. 12(1750)2020.PubMed/NCBI View Article : Google Scholar

61 

Jiang GM, Tan Y, Wang H, Peng L, Chen HT, Meng XJ, Li LL, Liu Y, Li WF and Shan H: The relationship between autophagy and the immune system and its applications for tumor immunotherapy. Mol Cancer. 18(17)2019.PubMed/NCBI View Article : Google Scholar

62 

Bolisetty S and Jaimes EA: Mitochondria and reactive oxygen species: Physiology and pathophysiology. Int J Mol Sci. 14:6306–6344. 2013.PubMed/NCBI View Article : Google Scholar

63 

Martinez J, Malireddi RS, Lu Q, Cunha LD, Pelletier S, Gingras S, Orchard R, Guan JL, Tan H, Peng J, et al: Molecular characterization of LC3-associated phagocytosis reveals distinct roles for Rubicon, NOX2, and autophagy proteins. Nat Cell Biol. 17:893–906. 2015.PubMed/NCBI View Article : Google Scholar

64 

Shin J, Song MH, Oh JW, Keum YS and Saini RK: Pro-oxidant actions of carotenoids in triggering apoptosis of cancer cells: A review of emerging evidence. Antioxidants (Basel). 9(532)2020.PubMed/NCBI View Article : Google Scholar

65 

Filteau SM, Rice AL, Ball JJ, Chakraborty J, Stoltzfus R, de Francisco A and Willumsen JF: Breast milk immune factors in Bangladeshi women supplemented postpartum with retinol or beta-carotene. Am J Clin Nutr. 69:953–958. 1999.PubMed/NCBI View Article : Google Scholar

66 

Kasperczyk S, Dobrakowski M, Kasperczyk J, Ostałowska A, Zalejska-Fiolka J and Birkner E: Beta-carotene reduces oxidative stress, improves glutathione metabolism and modifies antioxidant defense systems in lead-exposed workers. Toxicol Appl Pharmacol. 280:36–41. 2014.PubMed/NCBI View Article : Google Scholar

67 

Zbyradowski M, Duda M, Wisniewska-Becker A, Heriyanto Rajwa W, Fiedor J, Cvetkovic D, Pilch M and Fiedor L: Triplet-driven chemical reactivity of β-carotene and its biological implications. Nat Commun. 13(2474)2022.PubMed/NCBI View Article : Google Scholar

68 

Kim DK, Sim BR, Kim JI and Khang G: Functionalized silk fibroin film scaffold using β-Carotene for cornea endothelial cell regeneration. Colloids Surf B Biointerfaces. 164:340–346. 2018.PubMed/NCBI View Article : Google Scholar

69 

Kang H and Kim H: Astaxanthin and β-carotene in Helicobacter pylori-induced gastric inflammation: A mini-review on action mechanisms. J Cancer Prev. 22:57–61. 2017.PubMed/NCBI View Article : Google Scholar

70 

McGuire S: WHO guideline: Vitamin A supplementation in pregnant women. Geneva: WHO, 2011; WHO guideline: Vitamin A supplementation in postpartum women. Geneva: WHO, 2011. Adv Nutr. 3:215–216. 2012.PubMed/NCBI View Article : Google Scholar

71 

World Health Organization (WHO): Guideline: vitamin A supplementation in postpartum women. WHO, Geneva, 2011. https://apps.who.int/iris/handle/10665/44623.

72 

Tanaka T, Shnimizu M and Moriwaki H: Cancer chemoprevention by carotenoids. Molecules. 17:3202–3242. 2012.PubMed/NCBI View Article : Google Scholar

73 

Haskell MJ: The challenge to reach nutritional adequacy for vitamin A: β-carotene bioavailability and conversion-evidence in humans. Am J Clin Nutr. 96:1193S–1203S. 2012.PubMed/NCBI View Article : Google Scholar

74 

National Institutes of Health (NIH): Vitamin A and carotenoids. NIH, Bethesda, MD, pp1-19, 2022. https://ods.od.nih.gov/factsheets/VitaminA-HealthProfessional/. Accessed June 1, 2022.

75 

Xu J, Zhang J, Mao QF, Wu J and Wang Y: The interaction between autophagy and JAK/STAT3 signaling pathway in tumors. Front Genet. 13(880359)2022.PubMed/NCBI View Article : Google Scholar

76 

Kitakaze T, Harada N, Imagita H and Yamaji R: β-carotene increases muscle mass and hypertrophy in the soleus muscle in mice. J Nutr Sci Vitaminol (Tokyo). 61:481–487. 2015.PubMed/NCBI View Article : Google Scholar

77 

Geng N, Liu K, Lu J, Xu Y, Wang X, Wang R, Liu J, Liu Y and Han B: Autophagy of bovine mammary epithelial cell induced by intracellular Staphylococcus aureus. J Microbiol. 58:320–329. 2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

September-2022
Volume 17 Issue 3

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hasianna ST, Gunadi JW, Rohmawaty E and Lesmana R: Potential role of β‑carotene‑modulated autophagy in puerperal breast inflammation (Review). Biomed Rep 17: 75, 2022
APA
Hasianna, S.T., Gunadi, J.W., Rohmawaty, E., & Lesmana, R. (2022). Potential role of β‑carotene‑modulated autophagy in puerperal breast inflammation (Review). Biomedical Reports, 17, 75. https://doi.org/10.3892/br.2022.1558
MLA
Hasianna, S. T., Gunadi, J. W., Rohmawaty, E., Lesmana, R."Potential role of β‑carotene‑modulated autophagy in puerperal breast inflammation (Review)". Biomedical Reports 17.3 (2022): 75.
Chicago
Hasianna, S. T., Gunadi, J. W., Rohmawaty, E., Lesmana, R."Potential role of β‑carotene‑modulated autophagy in puerperal breast inflammation (Review)". Biomedical Reports 17, no. 3 (2022): 75. https://doi.org/10.3892/br.2022.1558