Next Article in Journal
Dyshidrosiform Bullous Pemphigoid
Previous Article in Journal
The p38/MK2 Axis in Monocytes of Fibromyalgia Syndrome Patients: An Explorative Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of Alpha-1 Antitrypsin in Type 1 and Type 2 Diabetes Mellitus

by
Sangmi S. Park
1,
Romy Rodriguez Ortega
1,
Christina W. Agudelo
1,
Jessica Perez Perez
1,
Brais Perez Gandara
1,
Itsaso Garcia-Arcos
1,
Cormac McCarthy
2 and
Patrick Geraghty
1,*
1
Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
2
University College Dublin School of Medicine, Education and Research Centre, St. Vincent’s University Hospital, D04 T6F4 Dublin, Ireland
*
Author to whom correspondence should be addressed.
Medicina 2021, 57(4), 397; https://doi.org/10.3390/medicina57040397
Submission received: 22 March 2021 / Revised: 12 April 2021 / Accepted: 17 April 2021 / Published: 20 April 2021
(This article belongs to the Section Endocrinology)

Abstract

:
Alpha-1 antitrypsin (AAT) has established anti-inflammatory and immunomodulatory effects in chronic obstructive pulmonary disease but there is increasing evidence of its role in other inflammatory and immune-mediated conditions, like diabetes mellitus (DM). AAT activity is altered in both developing and established type 1 diabetes mellitus (T1DM) as well in established type 2 DM (T2DM). Augmentation therapy with AAT appears to favorably impact T1DM development in mice models and to affect β-cell function and inflammation in humans with T1DM. The role of AAT in T2DM is less clear, but AAT activity appears to be reduced in T2DM. This article reviews these associations and emerging therapeutic strategies using AAT to treat DM.

1. Introduction

Alpha-1 antitrypsin (AAT) has established anti-inflammatory and immunomodulatory effects that go beyond its anti-protease activity, which are well documented in the pathogenesis of emphysema [1]. Although a large portion of the research in AAT has focused on pulmonary and liver diseases, there is a growing body of evidence linking AAT with other disease processes with an immune/inflammatory component [2,3,4,5]. Immune dysregulation and inflammation play a significant role in type 1 diabetes mellitus (T1DM) and is a subject of intense study for more than 100 years when Schmidt in 1902 described a peri-islet cellular infiltrate in the pancreas of a deceased 10-year-old child with diabetes [6,7,8,9]. Targeting immune/inflammatory pathways is a common and active area of research in the treatment of T1DM, which includes the use of agents such as glucocorticoids, cyclosporine, azathioprine, anti-thymocyte globulin, and rituximab but also cell therapies with varying results [10,11,12,13,14,15]. AAT’s association with diabetes mellitus started to become apparent in the 1980s when AAT activity was found to be reduced in patients with diabetes [16]. Since then, there is accumulating evidence of AAT’s role in T1DM [17,18,19] and a probable, but less clear role, in type 2 diabetes mellitus (T2DM) [20]. Here we review these associations and the potential for use of AAT targeted therapies as a strategy for the prevention and treatment of type 1 and type 2 diabetes mellitus.

2. Alpha-1 Antitrypsin Responses Play a Role in Type 1 Diabetes Mellitus

The AAT protein (encoded by the SERPINA1 gene) is primarily produced by the liver and circulated via the blood. AAT is an acute stress reactant protein and increases during stress conditions [21] and during the late stages of pregnancy [22]. AAT is primarily known as a potent anti-elastase protein [23]. AAT deficiency is extensively researched and many genetic variants of AAT are reported [24,25]. The most frequent and clinically significant alleles of AAT are PI*M, PI*S, and PI*Z, with PI*M being the normal/nonmutated allele [26]. The PI*S and PI*Z mutations account for approximately 95% of AAT deficiency cases. Carrying two copies of the severe PI*Z allele mutation leads to misfolding of the AAT protein, AAT protein retention in the liver, low serum levels of AAT (termed AAT deficiency) [27], and thus an inability to regulate inflammatory and proteolytic responses. Accumulation of the malformed protein in the liver leads to local damage [28]. Other clinical manifestations such as emphysema, panniculitis, and Wegener’s granulomatosis can occur in AAT deficient individuals [29]. Notably, AAT responses are significantly lower in the plasma of T1DM patients [16], and plasma anti-trypsin capability progressively decreases with a longer duration of diabetes [30]. Raising blood levels of AAT with augmentation therapy prevents T1DM development, prolongs islet allograft survival [18], increases insulin release capacity [31], and inhibits pancreatic B-cell apoptosis [32]. AAT treatment also significantly reduces HbA1c levels [33]. Importantly, expression of the human nonmutated AAT gene, by a recombinant adeno-associated virus, in nonobese diabetic (NOD) mice significantly reduced insulitis and prevented the development of overt hyperglycemia [17]. Serum AAT levels are significantly reduced in NOD mice [17] and AAT treatment expands the functional mass of the β cells in diabetic NOD mice [34], possibly due to changes in the inflammation milieu. Therefore, clinical and animal models suggest that AAT plays a major role in T1DM in addition to its established role in emphysema in AAT deficient individuals.

3. Alpha-1 Antitrypsin Might Have a Role in Type 2 Diabetes Mellitus Pathogenesis

Equally, another report highlighted an association of AAT deficiency with an increased risk of developing Type 2 diabetes (T2DM) [20]. High levels of degraded AAT are observed in the urine of T2DM patients with diabetic kidney disease [35]. T2DM often occurs with other comorbidities that include obesity and nonalcoholic fatty liver disease (NAFLD, or hepatic steatosis). In a clinical study, hepatic steatosis in the presence of acute pancreatitis resulted in reduced AAT levels that correlated with increased disease severity [36]. Alternatively, anti-trypsin capacity is lower in serum from gestational diabetes mellitus compared to healthy pregnant women [37]. However, additional data is needed on whether AAT has a direct role in T2DM pathogenesis and other types of diabetes.

4. What Mechanisms Could AAT Regulate in Preventing Diabetes Onset and Progression?

Despite AAT deficiency being extensively studied and exogenous AAT administered as a therapy to AAT-deficient emphysema subjects, little is known about the potential long-term effects of AAT signaling on T1DM patients. However, we do know several possible mechanisms that AAT signaling could mediate in preventing T1DM. AAT is primarily known to be an inhibitor of neutrophil protease, such as proteinase-1, elastase, thrombin, and trypsin [38]. In recent years, multiple roles of AAT beyond its anti-protease capacity have emerged [1,39], including activation of phosphatases [40], inhibition of caspase activity [41] and nitric oxide production [42], subduing HIV type 1 [43] and rhinovirus infectivity [44], reducing endoplasmic reticulum stress responses [45,46], regulating neutrophil degranulation [47,48], modifying dendritic cell maturation and promoting regulatory T cell (Treg) differentiation [49], increasing IL-10 and IL-1Ra release [50], minimizing epithelial barrier damage, and regulating IL-8-mediated neutrophil chemotaxis [51]. AAT also protects multiple proteins from undergoing proteolytic cleavage, such as phospholipid transporter protein (PLTP) [52]; thereby reducing lung inflammation and neutrophil degranulation [53]. AAT can counter damage associated with hypoxia [54,55,56,57], or in clinical circumstances with an underlying allogeneic background [58,59] or danger-associated molecular pattern agents (DAMPs) induced inflammation [60,61]. Equally, neutrophils isolated from human T1DM and T2DM subjects or mice are primed to produce neutrophil extracellular traps (NETs) [62], which could influence neutrophil responses in the lungs. Neutrophils from AAT deficient subjects have increased neutrophil responses [47,51,53] and AAT could counter elevated neutrophil NETs formation and degranulation. Therefore, the potential mechanisms for AAT’s protective role in T1DM could be vast.
Since dysfunctional inflammation is linked to the pathogenesis of T1DM, research was conducted on the beneficial effects of AAT in T1DM in terms of AAT’s anti-inflammatory properties. Indeed, AAT has anti-inflammatory and immune-modulating properties associated with enhancing pancreatic β-cell function, via regulation of IL-1β responses and the development of antigen-specific T regulatory cells [63]. AAT-treated mice have reduced serum TNF-α, lymphocytic infiltration, NF-κB activation, and JNK phosphorylation in their pancreatic β-cell islets [64]. One possible means of AAT regulating inflammation responses is through its regulation of cyclic adenosine monophosphate (cAMP). Increased insulin release capacity due to AAT stimulation coincides with elevated cAMP [31], as does AAT-induced phosphatase activity [40]. Elevated protein phosphatase activity following AAT stimulation attenuated cytokine and protease responses within the lungs [40]. AAT can also regulate nitric oxide responses, a known inducer of islet β cell death, by reducing its release [18]. Equally, AAT prevents apoptosis of pancreatic B-cells [32] and lung cells [65] by inhibiting caspase-3 activity. However, it must be noted that inflammation is not the only factor in T1DM and other AAT responses should be investigated.
The SERPINA1 gene is described to be a transcriptionally complex gene as it contains a number of different splicing events, including skipped exons, alternative donors, and alternative acceptors [66]. The SERPINA1 gene promotor becomes hypermethylated in the later term of pregnancy, leading to elevated levels of SERPINA1 expression [22]. AAT deficiency is suggested as a possible contributor to preterm premature rupture of membranes [67]. AAT protein expression levels are also controlled at the posttranscriptional level, by RNA structure influenced by noncoding gene regions [68]. It is also worth mentioning that there are several factors that could influence AAT activity rather than overall levels of gene expression and total plasma levels that could contribute to disease progression. Exposure to cigarette smoke can result in the oxidation of the methionine residues at sites 351 and 358 within the reactive center loop of the AAT protein, leading to diminished AAT anti-elastase function [69]. However, it was reported that AAT retains its anti-inflammatory properties even without its anti-elastase function [70]. Interestingly, serum levels of the AAT-oxidized low-density lipoprotein (LDL) complexes are high in smokers and decrease after smoking cessation due to weight gain [71] and could influence cardiovascular issues in emphysema patients [72]. AAT-LDL complexes also correlate with adiponectin levels in non-MetS subjects [73]. Circulating AAT undergoes glycation in hyperglycemic conditions and becomes inactivated [74]. The Z mutated form of AAT has increased fucosylation on N-glycans of Z-AAT that is associated with persistent inflammation [75]. AAT also circulates in high-density lipoprotein (HDL), as HDL was observed to be enriched with AAT and this may represent additional means for AAT to interact with many proteins [76]. AAT is secreted primarily from the liver and is distributed in plasma to many organs. However, where and when AAT incorporates into HDL is yet to be determined. AAT binds to unsaturated fatty acids, linoleic (C18:2) and oleic (C18:1), and the Z mutated form of AAT carries significant amounts of fatty acids [77]. This fatty acid-AAT complex induces expression of angiopoietin-like protein 4 (Angptl4) and fatty acid-binding protein 4 (FABP4), via a PPAR-dependent pathway [77], which could directly influence triacylglycerol homeostasis.
Finally, there is some mechanistic evidence suggesting that AAT could have a role in T2DM, with an imbalance between AAT and neutrophil elastase (NE) contributing to the development of obesity and insulin resistance in mice [78]. This was observed in NE knockout (Ela2−/−) mice and AAT transgenic mice, as they were resistant to high-fat diet-induced body weight gain, insulin resistance, inflammation, and fatty liver [78]. AAT regulated the AMP-activated protein kinase (AMPK) responses, fatty acid oxidation, and energy expenditure [78].

5. The Potential Use of AAT to Treat Diabetes

The only current safe treatment available for AAT deficiency-associated emphysema is intravenous AAT augmentation, which protects the lungs from disease progression [79]. On the basis of the potential benefits of AAT in ameliorating T1DM-associated inflammation and improving β-cell function, there are several clinical trials evaluating the potential for AAT therapy to treat T1DM. However, only a small number of these trials have published their findings and suggest certain populations of T1DM patients may benefit from AAT treatment (see Table 1). Small-sized studies in children and adults have demonstrated that AAT therapy is safe and well-tolerated in stage 3 T1DM [80] but a high dose may be required to see beneficial effects [81]. A recent phase II, double-blind, randomized, placebo-controlled clinical trial observed that AAT intervention reduced HbA1c levels in a subgroup of adolescents with recent-onset T1DM [82]. Children treated with AAT infusions have fewer IL-1β producing monocytes and dendritic cells [80].
Several animal and cell studies that explored AAT treatment reported AAT treatment reduces inflammation and retinal neurodegeneration via downregulation of NF-κB, iNOS, and TNF-α in the T1DM/Streptozotocin (STZ) mouse model of T1DM [83]. Equally, AAT can counter hyperglycemia-induced inflammation in retinal pigmented epithelial cells by controlling Akt responses [84]. In a rat insulinoma cell line, AAT increases insulin secretion in a glucose-dependent manner, while reducing TNF-α-induced apoptosis and cytokine production [31]. Recently, the application of CRISPR-Cas9 technology to correct AAT mutations in vivo was utilized to either correct an AAT mutation or enhance AAT production [85]. Equally, recombinant adeno-associated virus delivery of inducible human AAT significantly prevented T1DM development in NOD mice, and similar approaches could be utilized as future treatment approaches [86]. Therefore, AAT plays important roles in emphysema and T1DM, and possibly T2DM.

6. Conclusions

There is an emerging role of AAT in the onset and pathogenesis of diabetes mellitus especially T1DM and the use of AAT targeted therapies for the treatment of T1DM. These treatments could be effective on established T1DM but also might retard the progression of recent-onset T1DM. The role that AAT plays in T2DM pathogenesis is less clear and further research is needed to elucidate this association and possible therapeutic interventions.

Author Contributions

Conceptualization, S.S.P., R.R.O., C.M., and P.G.; writing—original draft preparation, S.S.P., R.R.O., C.M., and P.G.; writing—review and editing, S.S.P., R.R.O., C.W.A., J.P.P., B.P.G., I.G.-A., C.M., and P.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants made available to P.G. (the Alpha-1 Foundation (493373 and 614218).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bergin, D.A.; Hurley, K.; McElvaney, N.G.; Reeves, E.P. Alpha-1 antitrypsin: A potent anti-inflammatory and potential novel therapeutic agent. Arch. Immunol. Ther. Exp. 2012, 60, 81–97. [Google Scholar] [CrossRef] [PubMed]
  2. Breit, S.N.; Wakefield, D.; Robinson, J.P.; Luckhurst, E.; Clark, P.; Penny, R. The role of alpha 1-antitrypsin deficiency in the pathogenesis of immune disorders. Clin. Immunol. Immunopathol. 1985, 35, 363–380. [Google Scholar] [CrossRef]
  3. Pervakova, M.Y.; Emanuel, V.L.; Titova, O.N.; Lapin, S.V.; Mazurov, V.I.; Belyaeva, I.B.; Chudinov, A.L.; Blinova, T.V.; Surkova, E.A. The Diagnostic Value of Alpha-1-Antitrypsin Phenotype in Patients with Granulomatosis with Polyangiitis. Int. J. Rheumatol. 2016, 2016, 7831410. [Google Scholar] [CrossRef] [Green Version]
  4. McCarthy, C.; Orr, C.; Fee, L.T.; Carroll, T.P.; Dunlea, D.M.; Hunt, D.J.L.; Dunne, E.; O’Connell, P.; McCarthy, G.; Kenny, D.; et al. Brief Report: Genetic Variation of the alpha1-Antitrypsin Gene Is Associated With Increased Autoantibody Production in Rheumatoid Arthritis. Arthritis Rheumatol. 2017, 69, 1576–1579. [Google Scholar] [CrossRef]
  5. Franciosi, A.N.; Ralph, J.; O’Farrell, N.J.; Buckley, C.; Gulmann, C.; O’Kane, M.; Carroll, T.P.; McElvaney, N.G. Alpha-1 Antitrypsin Deficiency Associated Panniculitis. J. Am. Acad. Dermatol. 2021. [Google Scholar] [CrossRef]
  6. Bottazzo, G.F.; Cudworth, A.G.; Moul, D.J.; Doniach, D.; Festenstein, H. Evidence for a primary autoimmune type of diabetes mellitus. BMJ 1978, 2, 1253–1255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Bottazzo, G.F.; Florin-Christensen, A.; Doniach, D. Islet-cell antibodies in diabetes mellitus with autoimmune polyendocrine deficiencies. Lancet 1974, 2, 1279–1283. [Google Scholar] [CrossRef]
  8. von Meyenburg, H. Ueber “Insulitis” bei Diabetes. Schweiz Med. Wochenschr. 1940, 21, 554–557. [Google Scholar]
  9. Schmidt, M.B. Über die Beziehung der Langerhans’schen Inseln des Pankreas zum Diabetes mellitus. Münch Med. Wochenschr. 1902, 49, 51–54. [Google Scholar]
  10. Stiller, C.R.; Dupre, J.; Gent, M.; Jenner, M.R.; Keown, P.A.; Laupacis, A.; Martell, R.; Rodger, N.W.; von Graffenried, B.; Wolfe, B.M. Effects of cyclosporine immunosuppression in insulin-dependent diabetes mellitus of recent onset. Science 1984, 223, 1362–1367. [Google Scholar] [CrossRef]
  11. Chase, H.P.; Butler-Simon, N.; Garg, S.K.; Hayward, A.; Klingensmith, G.J.; Hamman, R.F.; O’Brien, D. Cyclosporine A for the treatment of new-onset insulin-dependent diabetes mellitus. Pediatrics 1990, 85, 241–245. [Google Scholar] [PubMed]
  12. Cook, J.J.; Hudson, I.; Harrison, L.C.; Dean, B.; Colman, P.G.; Werther, G.A.; Warne, G.L.; Court, J.M. Double-blind controlled trial of azathioprine in children with newly diagnosed type I diabetes. Diabetes 1989, 38, 779–783. [Google Scholar] [CrossRef] [PubMed]
  13. Eisenbarth, G.S.; Srikanta, S.; Jackson, R.; Rabinowe, S.; Dolinar, R.; Aoki, T.; Morris, M.A. Anti-thymocyte globulin and prednisone immunotherapy of recent onset type 1 diabetes mellitus. Diabetes Res. 1985, 2, 271–276. [Google Scholar] [PubMed]
  14. Pescovitz, M.D.; Greenbaum, C.J.; Krause-Steinrauf, H.; Becker, D.J.; Gitelman, S.E.; Goland, R.; Gottlieb, P.A.; Marks, J.B.; McGee, P.F.; Moran, A.M.; et al. Rituximab, B-lymphocyte depletion, and preservation of beta-cell function. N. Engl. J. Med. 2009, 361, 2143–2152. [Google Scholar] [CrossRef] [PubMed]
  15. Saudek, F.; Havrdova, T.; Boucek, P.; Karasova, L.; Novota, P.; Skibova, J. Polyclonal anti-T-cell therapy for type 1 diabetes mellitus of recent onset. Rev. Diabet. Stud. 2004, 1, 80–88. [Google Scholar] [CrossRef] [Green Version]
  16. Sandler, M.; Gemperli, B.M.; Hanekom, C.; Kuhn, S.H. Serum alpha 1-protease inhibitor in diabetes mellitus: Reduced concentration and impaired activity. Diabetes Res. Clin. Pract. 1988, 5, 249–255. [Google Scholar] [CrossRef]
  17. Lu, Y.; Tang, M.; Wasserfall, C.; Kou, Z.; Campbell-Thompson, M.; Gardemann, T.; Crawford, J.; Atkinson, M.; Song, S. Alpha1-antitrypsin gene therapy modulates cellular immunity and efficiently prevents type 1 diabetes in nonobese diabetic mice. Hum. Gene Ther. 2006, 17, 625–634. [Google Scholar] [CrossRef]
  18. Lewis, E.C.; Shapiro, L.; Bowers, O.J.; Dinarello, C.A. Alpha1-antitrypsin monotherapy prolongs islet allograft survival in mice. Proc. Natl. Acad. Sci. USA 2005, 102, 12153–12158. [Google Scholar] [CrossRef] [Green Version]
  19. Song, S.; Goudy, K.; Campbell-Thompson, M.; Wasserfall, C.; Scott-Jorgensen, M.; Wang, J.; Tang, Q.; Crawford, J.M.; Ellis, T.M.; Atkinson, M.A.; et al. Recombinant adeno-associated virus-mediated alpha-1 antitrypsin gene therapy prevents type I diabetes in NOD mice. Gene Ther. 2004, 11, 181–186. [Google Scholar] [CrossRef] [Green Version]
  20. Sandstrom, C.S.; Ohlsson, B.; Melander, O.; Westin, U.; Mahadeva, R.; Janciauskiene, S. An association between Type 2 diabetes and alpha-antitrypsin deficiency. Diabet. Med. 2008, 25, 1370–1373. [Google Scholar] [CrossRef]
  21. Perlmutter, D.H.; Schlesinger, M.J.; Pierce, J.A.; Punsal, P.I.; Schwartz, A.L. Synthesis of stress proteins is increased in individuals with homozygous PiZZ alpha 1-antitrypsin deficiency and liver disease. J. Clin. Investig. 1989, 84, 1555–1561. [Google Scholar] [CrossRef] [Green Version]
  22. Rotondo, J.C.; Oton-Gonzalez, L.; Selvatici, R.; Rizzo, P.; Pavasini, R.; Campo, G.C.; Lanzillotti, C.; Mazziotta, C.; De Mattei, M.; Tognon, M.; et al. SERPINA1 Gene Promoter Is Differentially Methylated in Peripheral Blood Mononuclear Cells of Pregnant Women. Front. Cell Dev. Biol. 2020, 8, 550543. [Google Scholar] [CrossRef]
  23. Morrison, H.M.; Kramps, J.A.; Burnett, D.; Stockley, R.A. Lung lavage fluid from patients with alpha 1-proteinase inhibitor deficiency or chronic obstructive bronchitis: Anti-elastase function and cell profile. Clin. Sci. 1987, 72, 373–381. [Google Scholar] [CrossRef]
  24. de Serres, F.J.; Blanco, I.; Fernández-Bustillo, E. Ethnic differences in alpha-1 antitrypsin deficiency in the United States of America. Ther. Adv. Respir. Dis. 2010, 4, 63–70. [Google Scholar] [CrossRef]
  25. Abboud, R.T.; Nelson, T.N.; Jung, B.; Mattman, A. Alpha1-antitrypsin deficiency: A clinical-genetic overview. Appl. Clin. Genet. 2011, 4, 55–65. [Google Scholar] [CrossRef] [Green Version]
  26. Seixas, S.; Marques, P.I. Known Mutations at the Cause of Alpha-1 Antitrypsin Deficiency an Updated Overview of SERPINA1 Variation Spectrum. Appl. Clin. Genet. 2021, 14, 173–194. [Google Scholar] [CrossRef] [PubMed]
  27. Ogushi, F.; Fells, G.A.; Hubbard, R.C.; Straus, S.D.; Crystal, R.G. Z-type alpha 1-antitrypsin is less competent than M1-type alpha 1-antitrypsin as an inhibitor of neutrophil elastase. J. Clin. Investig. 1987, 80, 1366–1374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Lomas, D.A.; Evans, D.L.; Finch, J.T.; Carrell, R.W. The mechanism of Z alpha 1-antitrypsin accumulation in the liver. Nature 1992, 357, 605–607. [Google Scholar] [CrossRef]
  29. Greene, C.M.; Marciniak, S.J.; Teckman, J.; Ferrarotti, I.; Brantly, M.L.; Lomas, D.A.; Stoller, J.K.; McElvaney, N.G. alpha1-Antitrypsin deficiency. Nat. Rev. Dis. Primers 2016, 2, 16051. [Google Scholar] [CrossRef] [PubMed]
  30. Hashemi, M.; Naderi, M.; Rashidi, H.; Ghavami, S. Impaired activity of serum alpha-1-antitrypsin in diabetes mellitus. Diabetes Res. Clin. Pract. 2007, 75, 246–248. [Google Scholar] [CrossRef]
  31. Kalis, M.; Kumar, R.; Janciauskiene, S.; Salehi, A.; Cilio, C.M. alpha 1-antitrypsin enhances insulin secretion and prevents cytokine-mediated apoptosis in pancreatic beta-cells. Islets 2010, 2, 185–189. [Google Scholar] [CrossRef] [Green Version]
  32. Zhang, B.; Lu, Y.; Campbell-Thompson, M.; Spencer, T.; Wasserfall, C.; Atkinson, M.; Song, S. Alpha1-antitrypsin protects beta-cells from apoptosis. Diabetes 2007, 56, 1316–1323. [Google Scholar] [CrossRef] [Green Version]
  33. Rachmiel, M.; Strauss, P.; Dror, N.; Benzaquen, H.; Horesh, O.; Tov, N.; Weintrob, N.; Landau, Z.; Ben-Ami, M.; Haim, A.; et al. Alpha-1 antitrypsin therapy is safe and well tolerated in children and adolescents with recent onset type 1 diabetes mellitus. Pediatr. Diabetes 2016, 17, 351–359. [Google Scholar] [CrossRef]
  34. Koulmanda, M.; Bhasin, M.; Hoffman, L.; Fan, Z.; Qipo, A.; Shi, H.; Bonner-Weir, S.; Putheti, P.; Degauque, N.; Libermann, T.A.; et al. Curative and beta cell regenerative effects of alpha1-antitrypsin treatment in autoimmune diabetic NOD mice. Proc. Natl. Acad. Sci. USA 2008, 105, 16242–16247. [Google Scholar] [CrossRef] [Green Version]
  35. Brondani, L.A.; Soares, A.A.; Recamonde-Mendoza, M.; Dall’Agnol, A.; Camargo, J.L.; Monteiro, K.M.; Silveiro, S.P. Urinary peptidomics and bioinformatics for the detection of diabetic kidney disease. Sci. Rep. 2020, 10, 1242. [Google Scholar] [CrossRef] [Green Version]
  36. Wang, Q.; Du, J.; Yu, P.; Bai, B.; Zhao, Z.; Wang, S.; Zhu, J.; Feng, Q.; Gao, Y.; Zhao, Q.; et al. Hepatic steatosis depresses alpha-1-antitrypsin levels in human and rat acute pancreatitis. Sci. Rep. 2015, 5, 17833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Yaghmaei, M.; Hashemi, M.; Shikhzadeh, A.; Mokhtari, M.; Niazi, A.; Ghavami, S. Serum trypsin inhibitory capacity in normal pregnancy and gestational diabetes mellitus. Diabetes Res. Clin. Pract. 2009, 84, 201–204. [Google Scholar] [CrossRef]
  38. Korkmaz, B.; Horwitz, M.S.; Jenne, D.E.; Gauthier, F. Neutrophil elastase, proteinase 3, and cathepsin G as therapeutic targets in human diseases. Pharmacol. Rev. 2010, 62, 726–759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Hunt, J.M.; Tuder, R. Alpha 1 anti-trypsin: One protein, many functions. Curr. Mol. Med. 2012, 12, 827–835. [Google Scholar] [CrossRef] [PubMed]
  40. Geraghty, P.; Eden, E.; Pillai, M.; Campos, M.; McElvaney, N.G.; Foronjy, R.F. alpha1-Antitrypsin activates protein phosphatase 2A to counter lung inflammatory responses. Am. J. Respir. Crit. Care Med. 2014, 190, 1229–1242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Petrache, I.; Fijalkowska, I.; Medler, T.R.; Skirball, J.; Cruz, P.; Zhen, L.; Petrache, H.I.; Flotte, T.R.; Tuder, R.M. alpha-1 antitrypsin inhibits caspase-3 activity, preventing lung endothelial cell apoptosis. Am. J. Pathol. 2006, 169, 1155–1166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Chan, E.D.; Pott, G.B.; Silkoff, P.E.; Ralston, A.H.; Bryan, C.L.; Shapiro, L. Alpha-1-antitrypsin inhibits nitric oxide production. J. Leukoc. Biol. 2012, 92, 1251–1260. [Google Scholar] [CrossRef] [PubMed]
  43. Shapiro, L.; Pott, G.B.; Ralston, A.H. Alpha-1-antitrypsin inhibits human immunodeficiency virus type 1. FASEB J. 2001, 15, 115–122. [Google Scholar] [CrossRef] [PubMed]
  44. Berman, R.; Jiang, D.; Wu, Q.; Chu, H.W. alpha1-Antitrypsin reduces rhinovirus infection in primary human airway epithelial cells exposed to cigarette smoke. Int. J. Chron. Obstruct. Pulmon Dis. 2016, 11, 1279–1286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Carlson, J.A.; Rogers, B.B.; Sifers, R.N.; Finegold, M.J.; Clift, S.M.; DeMayo, F.J.; Bullock, D.W.; Woo, S.L. Accumulation of PiZ alpha 1-antitrypsin causes liver damage in transgenic mice. J. Clin. Investig. 1989, 83, 1183–1190. [Google Scholar] [CrossRef] [Green Version]
  46. Carroll, T.P.; Greene, C.M.; O’Connor, C.A.; Nolan, A.M.; O’Neill, S.J.; McElvaney, N.G. Evidence for unfolded protein response activation in monocytes from individuals with alpha-1 antitrypsin deficiency. J. Immunol. 2010, 184, 4538–4546. [Google Scholar] [CrossRef] [Green Version]
  47. Bergin, D.A.; Reeves, E.P.; Hurley, K.; Wolfe, R.; Jameel, R.; Fitzgerald, S.; McElvaney, N.G. The circulating proteinase inhibitor alpha-1 antitrypsin regulates neutrophil degranulation and autoimmunity. Sci. Transl. Med. 2014, 6, 217ra211. [Google Scholar] [CrossRef]
  48. Mueller, C.; Gernoux, G.; Gruntman, A.M.; Borel, F.; Reeves, E.P.; Calcedo, R.; Rouhani, F.N.; Yachnis, A.; Humphries, M.; Campbell-Thompson, M.; et al. 5 Year Expression and Neutrophil Defect Repair after Gene Therapy in Alpha-1 Antitrypsin Deficiency. Mol. Ther. 2017, 25, 1387–1394. [Google Scholar] [CrossRef] [Green Version]
  49. Shahaf, G.; Moser, H.; Ozeri, E.; Mizrahi, M.; Abecassis, A.; Lewis, E.C. alpha-1-antitrypsin gene delivery reduces inflammation, increases T-regulatory cell population size and prevents islet allograft rejection. Mol. Med. 2011, 17, 1000–1011. [Google Scholar] [CrossRef]
  50. Abecassis, A.; Schuster, R.; Shahaf, G.; Ozeri, E.; Green, R.; Ochayon, D.E.; Rider, P.; Lewis, E.C. alpha1-antitrypsin increases interleukin-1 receptor antagonist production during pancreatic islet graft transplantation. Cell Mol. Immunol. 2014, 11, 377–386. [Google Scholar] [CrossRef] [Green Version]
  51. Bergin, D.A.; Reeves, E.P.; Meleady, P.; Henry, M.; McElvaney, O.J.; Carroll, T.P.; Condron, C.; Chotirmall, S.H.; Clynes, M.; O’Neill, S.J.; et al. alpha-1 Antitrypsin regulates human neutrophil chemotaxis induced by soluble immune complexes and IL-8. J. Clin. Investig. 2010, 120, 4236–4250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Brehm, A.; Geraghty, P.; Campos, M.; Garcia-Arcos, I.; Dabo, A.J.; Gaffney, A.; Eden, E.; Jiang, X.C.; D’Armiento, J.; Foronjy, R. Cathepsin G degradation of phospholipid transfer protein (PLTP) augments pulmonary inflammation. FASEB J. 2014, 28, 2318–2331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ochieng, P.; Nath, S.; Macarulay, R.; Eden, E.; Dabo, A.; Campos, M.; Jiang, X.C.; Foronjy, R.F.; Geraghty, P. Phospholipid transfer protein and alpha-1 antitrypsin regulate Hck kinase activity during neutrophil degranulation. Sci. Rep. 2018, 8, 15394. [Google Scholar] [CrossRef] [PubMed]
  54. Daemen, M.A.; Heemskerk, V.H.; van’t Veer, C.; Denecker, G.; Wolfs, T.G.; Vandenabeele, P.; Buurman, W.A. Functional protection by acute phase proteins alpha(1)-acid glycoprotein and alpha(1)-antitrypsin against ischemia/reperfusion injury by preventing apoptosis and inflammation. Circulation 2000, 102, 1420–1426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Toldo, S.; Seropian, I.M.; Mezzaroma, E.; Van Tassell, B.W.; Salloum, F.N.; Lewis, E.C.; Voelkel, N.; Dinarello, C.A.; Abbate, A. Alpha-1 antitrypsin inhibits caspase-1 and protects from acute myocardial ischemia-reperfusion injury. J. Mol. Cell Cardiol. 2011, 51, 244–251. [Google Scholar] [CrossRef]
  56. Gao, W.; Zhao, J.; Kim, H.; Xu, S.; Chen, M.; Bai, X.; Toba, H.; Cho, H.R.; Zhang, H.; Keshavjeel, S.; et al. alpha1-Antitrypsin inhibits ischemia reperfusion-induced lung injury by reducing inflammatory response and cell death. J. Heart Lung Transplant. 2014, 33, 309–315. [Google Scholar] [CrossRef]
  57. Moldthan, H.L.; Hirko, A.C.; Thinschmidt, J.S.; Grant, M.B.; Li, Z.; Peris, J.; Lu, Y.; Elshikha, A.S.; King, M.A.; Hughes, J.A.; et al. Alpha 1-antitrypsin therapy mitigated ischemic stroke damage in rats. J. Stroke Cerebrovasc. Dis. 2014, 23, e355–e363. [Google Scholar] [CrossRef] [Green Version]
  58. Marcondes, A.M.; Li, X.; Tabellini, L.; Bartenstein, M.; Kabacka, J.; Sale, G.E.; Hansen, J.A.; Dinarello, C.A.; Deeg, H.J. Inhibition of IL-32 activation by alpha-1 antitrypsin suppresses alloreactivity and increases survival in an allogeneic murine marrow transplantation model. Blood 2011, 118, 5031–5039. [Google Scholar] [CrossRef] [Green Version]
  59. Tawara, I.; Sun, Y.; Lewis, E.C.; Toubai, T.; Evers, R.; Nieves, E.; Azam, T.; Dinarello, C.A.; Reddy, P. Alpha-1-antitrypsin monotherapy reduces graft-versus-host disease after experimental allogeneic bone marrow transplantation. Proc. Natl. Acad. Sci. USA 2012, 109, 564–569. [Google Scholar] [CrossRef] [Green Version]
  60. Finotti, P.; Pagetta, A. A heat shock protein70 fusion protein with alpha1-antitrypsin in plasma of type 1 diabetic subjects. Biochem. Biophys. Res. Commun. 2004, 315, 297–305. [Google Scholar] [CrossRef]
  61. Ochayon, D.E.; Mizrahi, M.; Shahaf, G.; Baranovski, B.M.; Lewis, E.C. Human alpha1-Antitrypsin Binds to Heat-Shock Protein gp96 and Protects from Endogenous gp96-Mediated Injury In vivo. Front. Immunol. 2013, 4, 320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Wong, S.L.; Demers, M.; Martinod, K.; Gallant, M.; Wang, Y.; Goldfine, A.B.; Kahn, C.R.; Wagner, D.D. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat. Med. 2015, 21, 815–819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Lewis, E.C.; Mizrahi, M.; Toledano, M.; Defelice, N.; Wright, J.L.; Churg, A.; Shapiro, L.; Dinarello, C.A. alpha1-Antitrypsin monotherapy induces immune tolerance during islet allograft transplantation in mice. Proc. Natl. Acad. Sci. USA 2008, 105, 16236–16241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Wang, J.; Sun, Z.; Gou, W.; Adams, D.B.; Cui, W.; Morgan, K.A.; Strange, C.; Wang, H. alpha-1 Antitrypsin Enhances Islet Engraftment by Suppression of Instant Blood-Mediated Inflammatory Reaction. Diabetes 2017, 66, 970–980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Petrache, I.; Fijalkowska, I.; Zhen, L.; Medler, T.R.; Brown, E.; Cruz, P.; Choe, K.H.; Taraseviciene-Stewart, L.; Scerbavicius, R.; Shapiro, L.; et al. A novel antiapoptotic role for alpha1-antitrypsin in the prevention of pulmonary emphysema. Am. J. Respir. Crit. Care Med. 2006, 173, 1222–1228. [Google Scholar] [CrossRef] [Green Version]
  66. Lackey, L.; McArthur, E.; Laederach, A. Increased Transcript Complexity in Genes Associated with Chronic Obstructive Pulmonary Disease. PLoS ONE 2015, 10, e0140885. [Google Scholar] [CrossRef]
  67. Baron, J.; Sheiner, E.; Abecassis, A.; Ashkenazi, E.; Shahaf, G.; Salem, S.Y.; Madar, T.; Twina, G.; Wiznitzer, A.; Holcberg, G.; et al. alpha1-antitrypsin insufficiency is a possible contributor to preterm premature rupture of membranes. J. Matern. Fetal Neonatal Med. 2012, 25, 934–937. [Google Scholar] [CrossRef]
  68. Corley, M.; Solem, A.; Phillips, G.; Lackey, L.; Ziehr, B.; Vincent, H.A.; Mustoe, A.M.; Ramos, S.B.V.; Weeks, K.M.; Moorman, N.J.; et al. An RNA structure-mediated, posttranscriptional model of human alpha-1-antitrypsin expression. Proc. Natl. Acad. Sci. USA 2017, 114, E10244–E10253. [Google Scholar] [CrossRef] [Green Version]
  69. Taggart, C.; Cervantes-Laurean, D.; Kim, G.; McElvaney, N.G.; Wehr, N.; Moss, J.; Levine, R.L. Oxidation of either methionine 351 or methionine 358 in alpha 1-antitrypsin causes loss of anti-neutrophil elastase activity. J. Biol. Chem. 2000, 275, 27258–27265. [Google Scholar] [CrossRef]
  70. Jonigk, D.; Al-Omari, M.; Maegel, L.; Muller, M.; Izykowski, N.; Hong, J.; Hong, K.; Kim, S.H.; Dorsch, M.; Mahadeva, R.; et al. Anti-inflammatory and immunomodulatory properties of alpha1-antitrypsin without inhibition of elastase. Proc. Natl. Acad. Sci. USA 2013, 110, 15007–15012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Komiyama, M.; Wada, H.; Ura, S.; Yamakage, H.; Satoh-Asahara, N.; Shimada, S.; Akao, M.; Koyama, H.; Kono, K.; Shimatsu, A.; et al. The effects of weight gain after smoking cessation on atherogenic alpha1-antitrypsin-low-density lipoprotein. Heart Vessels 2015, 30, 734–739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Funamoto, M.; Sunagawa, Y.; Katanasaka, Y.; Miyazaki, Y.; Imaizumi, A.; Kakeya, H.; Yamakage, H.; Satoh-Asahara, N.; Komiyama, M.; Wada, H.; et al. Highly absorptive curcumin reduces serum atherosclerotic low-density lipoprotein levels in patients with mild COPD. Int. J. Chron. Obstruct. Pulmon Dis. 2016, 11, 2029–2034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Kotani, K.; Yamada, T.; Taniguchi, N. The association between adiponectin, HDL-cholesterol and alpha1-antitrypsin-LDL in female subjects without metabolic syndrome. Lipids Health Dis. 2010, 9, 147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Austin, G.E.; Mullins, R.H.; Morin, L.G. Non-enzymic glycation of individual plasma proteins in normoglycemic and hyperglycemic patients. Clin. Chem. 1987, 33, 2220–2224. [Google Scholar] [CrossRef] [PubMed]
  75. McCarthy, C.; Saldova, R.; O’Brien, M.E.; Bergin, D.A.; Carroll, T.P.; Keenan, J.; Meleady, P.; Henry, M.; Clynes, M.; Rudd, P.M.; et al. Increased outer arm and core fucose residues on the N-glycans of mutated alpha-1 antitrypsin protein from alpha-1 antitrypsin deficient individuals. J. Proteome Res. 2014, 13, 596–605. [Google Scholar] [CrossRef]
  76. Moreno, J.A.; Ortega-Gomez, A.; Rubio-Navarro, A.; Louedec, L.; Ho-Tin-Noe, B.; Caligiuri, G.; Nicoletti, A.; Levoye, A.; Plantier, L.; Meilhac, O. High-density lipoproteins potentiate alpha1-antitrypsin therapy in elastase-induced pulmonary emphysema. Am. J. Respir. Cell Mol. Biol. 2014, 51, 536–549. [Google Scholar] [CrossRef]
  77. Frenzel, E.; Wrenger, S.; Brugger, B.; Salipalli, S.; Immenschuh, S.; Aggarwal, N.; Lichtinghagen, R.; Mahadeva, R.; Marcondes, A.M.; Dinarello, C.A.; et al. alpha1-Antitrypsin Combines with Plasma Fatty Acids and Induces Angiopoietin-like Protein 4 Expression. J. Immunol. 2015, 195, 3605–3616. [Google Scholar] [CrossRef] [Green Version]
  78. Mansuy-Aubert, V.; Zhou, Q.L.; Xie, X.; Gong, Z.; Huang, J.Y.; Khan, A.R.; Aubert, G.; Candelaria, K.; Thomas, S.; Shin, D.J.; et al. Imbalance between neutrophil elastase and its inhibitor alpha1-antitrypsin in obesity alters insulin sensitivity, inflammation, and energy expenditure. Cell Metab. 2013, 17, 534–548. [Google Scholar] [CrossRef] [Green Version]
  79. Chapman, K.R.; Burdon, J.G.; Piitulainen, E.; Sandhaus, R.A.; Seersholm, N.; Stocks, J.M.; Stoel, B.C.; Huang, L.; Yao, Z.; Edelman, J.M.; et al. Intravenous augmentation treatment and lung density in severe alpha1 antitrypsin deficiency (RAPID): A randomised, double-blind, placebo-controlled trial. Lancet 2015, 386, 360–368. [Google Scholar] [CrossRef]
  80. Gottlieb, P.A.; Alkanani, A.K.; Michels, A.W.; Lewis, E.C.; Shapiro, L.; Dinarello, C.A.; Zipris, D. alpha1-Antitrypsin therapy downregulates toll-like receptor-induced IL-1beta responses in monocytes and myeloid dendritic cells and may improve islet function in recently diagnosed patients with type 1 diabetes. J. Clin. Endocrinol. Metab. 2014, 99, E1418–E1426. [Google Scholar] [CrossRef]
  81. Weir, G.C.; Ehlers, M.R.; Harris, K.M.; Kanaparthi, S.; Long, A.; Phippard, D.; Weiner, L.J.; Jepson, B.; McNamara, J.G.; Koulmanda, M.; et al. Alpha-1 antitrypsin treatment of new-onset type 1 diabetes: An open-label, phase I clinical trial (RETAIN) to assess safety and pharmacokinetics. Pediatr. Diabetes 2018, 19, 945–954. [Google Scholar] [CrossRef] [PubMed]
  82. Lebenthal, Y.; Brener, A.; Hershkovitz, E.; Shehadeh, N.; Shalitin, S.; Lewis, E.C.; Elias, D.; Haim, A.; Barash, G.; Loewenthal, N.; et al. A Phase II, Double-Blind, Randomized, Placebo-Controlled, Multicenter Study Evaluating the Efficacy and Safety of Alpha-1 Antitrypsin (AAT) (Glassia((R))) in the Treatment of Recent-Onset Type 1 Diabetes. Int. J. Mol. Sci. 2019, 20, 6032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Ortiz, G.; Lopez, E.S.; Salica, J.P.; Potilinski, C.; Fernandez Acquier, M.; Chuluyan, E.; Gallo, J.E. Alpha-1-antitrypsin ameliorates inflammation and neurodegeneration in the diabetic mouse retina. Exp. Eye Res. 2018, 174, 29–39. [Google Scholar] [CrossRef] [PubMed]
  84. Potilinski, M.C.; Ortiz, G.A.; Salica, J.P.; Lopez, E.S.; Fernandez Acquier, M.; Chuluyan, E.; Gallo, J.E. Elucidating the mechanism of action of alpha-1-antitrypsin using retinal pigment epithelium cells exposed to high glucose. Potential use in diabetic retinopathy. PLoS ONE 2020, 15, e0228895. [Google Scholar] [CrossRef] [PubMed]
  85. Song, C.Q.; Wang, D.; Jiang, T.; O’Connor, K.; Tang, Q.; Cai, L.; Li, X.; Weng, Z.; Yin, H.; Gao, G.; et al. In Vivo Genome Editing Partially Restores Alpha1-Antitrypsin in a Murine Model of AAT Deficiency. Hum. Gene Ther. 2018, 29, 853–860. [Google Scholar] [CrossRef] [PubMed]
  86. Ma, H.; Lu, Y.; Lowe, K.; van der Meijden-Erkelens, L.; Wasserfall, C.; Atkinson, M.A.; Song, S. Regulated hAAT Expression from a Novel rAAV Vector and Its Application in the Prevention of Type 1 Diabetes. J. Clin. Med. 2019, 8, 1321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Summary of clinical trials of Alpha-1 Antitrypsin in type 1 diabetes mellitus.
Table 1. Summary of clinical trials of Alpha-1 Antitrypsin in type 1 diabetes mellitus.
StudyDesignPopulationInterventionMain Outcomes
Gottlieb, P.A. 2014 [80]Prospective, phase I, open-label interventional trial.N = 12 (subjects with T1DM within ∼4 years from disease diagnosis.)
Age: 24.6 ± 10.5 years (range, 12–39 years)
Sex: 4 females and 8 males.
8 consecutive weekly infusions of 80 mg/kg of AAT (Aralast; Baxter Inc) were given.
  • No significant adverse effects were detected.
  • Decreased total content of TLR4-induced cellular IL-1β.
  • Improved β-cell function correlated with lower IL-1β production.
Rachmiel, M. 2016 [33]Prospective, phase I/II open-label, interventional trial.n = 24 (recently diagnosed subjects with T1DM
Age: 12.9 ± 2.4 years (range, 9.8–17.6)
Sex: 12 females and 12 males.
18 infusions of 40, 60, or 80 mg/kg/dose high-purity, liquid, ready to use AAT (Glassia®; Kamada Ltd.) over 28 weeks.
12 weeks: weekly. 12–20 weeks: once every 2 weeks,
20–28 weeks: once every 4 weeks
  • No serious adverse events were reported.
  • Glycemic control parameters improved during the study in all groups, independent of dosage.
  • Eight subjects (33.3%) that were considered possible responders had a shorter duration of T1DM) and a greater decrease in their HbA1c.
Weir, G.C. 2018 [66]Prospective, phase I multicenter, open-label, dose-escalation study
(RETAIN).
n = 16 (within 100 days of diagnosis of T1DM)
Age: 8 adults aged 16 to 35 years and, 8 children aged 8 to 15 years)
12 infusions of AAT (Aralast NP; Baxter Inc): a low dose of 45 mg/kg weekly for 6 weeks, followed by a higher dose of 90 mg/kg for 6 weeks.
  • C-peptide secretion during a mixed meal remained relatively stable during the treatment period in adults and decreased in children.
  • HbA1c and Insulin usage remained relatively stable during the treatment period on both groups but gradually increased afterward.
  • AAT suppressed expression of genes involved in NF-κB activation and apoptosis pathways.
Lebenthal, Y. 2019 [82]Phase II, Double-Blind, Randomized, Placebo-Controlled, Multicenter Studyn = 69 (recently diagnosed T1DM patients)
Age: 13.1 ± 4.1 years
Sex: 32 females and 37 males.
22 infusions of AAT (Glassia®; Kamada Ltd.) (60 or 120 mg/kg) or placebo.
  • AAT was tolerated well, with a similar safety profile between groups.
  • C-peptide, glycated hemoglobin (HbA1c), and the total insulin dose (U/kg) were similar across groups.
  • C-peptide AUC levels in the AAT-120 mg/kg adolescent group remained relatively stable in contrast to the decline observed in the placebo and AAT-60 mg/kg groups.
  • The frequency of responders with at least 95% β-cell function reserve was 29% in the AAT-120 group and nil in the placebo.
AAT: alpha-1 antitrypsin; T1DM: type 1 diabetes mellitus; TLR-4: Toll-like receptor 4; IL-1β: interleukin-1 β, HbA1c: glycated hemoglobin A1c; NF-κB: nuclear factor-kappa B; AUC: area under the curve.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Park, S.S.; Rodriguez Ortega, R.; Agudelo, C.W.; Perez Perez, J.; Perez Gandara, B.; Garcia-Arcos, I.; McCarthy, C.; Geraghty, P. Therapeutic Potential of Alpha-1 Antitrypsin in Type 1 and Type 2 Diabetes Mellitus. Medicina 2021, 57, 397. https://doi.org/10.3390/medicina57040397

AMA Style

Park SS, Rodriguez Ortega R, Agudelo CW, Perez Perez J, Perez Gandara B, Garcia-Arcos I, McCarthy C, Geraghty P. Therapeutic Potential of Alpha-1 Antitrypsin in Type 1 and Type 2 Diabetes Mellitus. Medicina. 2021; 57(4):397. https://doi.org/10.3390/medicina57040397

Chicago/Turabian Style

Park, Sangmi S., Romy Rodriguez Ortega, Christina W. Agudelo, Jessica Perez Perez, Brais Perez Gandara, Itsaso Garcia-Arcos, Cormac McCarthy, and Patrick Geraghty. 2021. "Therapeutic Potential of Alpha-1 Antitrypsin in Type 1 and Type 2 Diabetes Mellitus" Medicina 57, no. 4: 397. https://doi.org/10.3390/medicina57040397

Article Metrics

Back to TopTop