Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Regina Irwin is active.

Publication


Featured researches published by Regina Irwin.


Journal of Cellular Physiology | 2014

Probiotic L. reuteri Treatment Prevents Bone Loss in a Menopausal Ovariectomized Mouse Model

Robert A. Britton; Regina Irwin; Darin Quach; Laura Schaefer; Jing Zhang; Taehyung Lee; Narayanan Parameswaran; Laura R. McCabe

Estrogen deficiency is a major risk factor for osteoporosis that is associated with bone inflammation and resorption. Half of women over the age of 50 will experience an osteoporosis related fracture in their lifetime, thus novel therapies are needed to combat post‐menopausal bone loss. Recent studies suggest an important role for gut‐bone signaling pathways and the microbiota in regulating bone health. Given that the bacterium Lactobacillus reuteri ATCC PTA 6475 (L. reuteri) secretes beneficial immunomodulatory factors, we examined if this candidate probiotic could reduce bone loss associated with estrogen deficiency in an ovariectomized (Ovx) mouse menopausal model. Strikingly, L. reuteri treatment significantly protected Ovx mice from bone loss. Osteoclast bone resorption markers and activators (Trap5 and RANKL) as well as osteoclastogenesis are significantly decreased in L. reuteri‐treated mice. Consistent with this, L. reuteri suppressed Ovx‐induced increases in bone marrow CD4+ T‐lymphocytes (which promote osteoclastogenesis) and directly suppressed osteoclastogenesis in vitro. We also identified that L. reuteri treatment modifies microbial communities in the Ovx mouse gut. Together, our studies demonstrate that L. reuteri treatment suppresses bone resorption and loss associated with estrogen deficiency. Thus, L. reuteri treatment may be a straightforward and cost‐effective approach to reduce post‐menopausal bone loss. J. Cell. Physiol. 229: 1822–1830, 2014.


Journal of Cellular Physiology | 2011

The bone marrow microenvironment contributes to type I diabetes induced osteoblast death

Lindsay M. Coe; Regina Irwin; Dennean Lippner; Laura R. McCabe

Type I diabetes increases an individuals risk for bone loss and fracture, predominantly through suppression of osteoblast activity (bone formation). During diabetes onset, levels of blood glucose and pro‐inflammatory cytokines (including tumor necrosis factor α (TNFα)) increased. At the same time, levels of osteoblast markers are rapidly decreased and stay decreased chronically (i.e., 40 days later) at which point bone loss is clearly evident. We hypothesized that early bone marrow inflammation can promote osteoblast death and hence reduced osteoblast markers. Indeed, examination of type I diabetic mouse bones demonstrates a greater than twofold increase in osteoblast TUNEL staining and increased expression of pro‐apoptotic factors. Osteoblast death was amplified in both pharmacologic and spontaneous diabetic mouse models. Given the known signaling and inter‐relationships between marrow cells and osteoblasts, we examined the role of diabetic marrow in causing the osteoblast death. Co‐culture studies demonstrate that compared to control marrow cells, diabetic bone marrow cells increase osteoblast (MC3T3 and bone marrow derived) caspase 3 activity and the ratio of Bax/Bcl‐2 expression. Mouse blood glucose levels positively correlated with bone marrow induced osteoblast death and negatively correlated with osteocalcin expression in bone, suggesting a relationship between type I diabetes, bone marrow and osteoblast death. TNF expression was elevated in diabetic marrow (but not co‐cultured osteoblasts); therefore, we treated co‐cultures with TNFα neutralizing antibodies. The antibody protected osteoblasts from bone marrow induced death. Taken together, our findings implicate the bone marrow microenvironment and TNFα in mediating osteoblast death and contributing to type I diabetic bone loss. J. Cell. Physiol. 226: 477–483, 2011.


Journal of Cellular Physiology | 2009

Bone inflammation and altered gene expression with type I diabetes early onset

Katherine J. Motyl; Sergiu Botolin; Regina Irwin; Daniel M. Appledorn; Tejas B. Kadakia; Andrea Amalfitano; Richard C. Schwartz; Laura R. McCabe

Type I diabetes is associated with bone loss and marrow adiposity. To identify early events involved in the etiology of diabetic bone loss, diabetes was induced in mice by multiple low dose streptozotocin injections. Serum markers of bone metabolism and inflammation as well as tibial gene expression were examined between 1 and 17 days post‐injection (dpi). At 3 dpi, when blood glucose levels were significantly elevated, body, fat pad and muscle mass were decreased. Serum markers of bone resorption and formation significantly decreased at 5 dpi in diabetic mice and remained suppressed throughout the time course. An osteoclast gene, TRAP5 mRNA, was suppressed at early and late time points. Suppression of osteogenic genes (runx2 and osteocalcin) and induction of adipogenic genes (PPARγ2 and aP2) were evident as early as 5 dpi. These changes were associated with an elevation of serum cytokines, but more importantly we observed an increase in the expression of cytokines in bone, supporting the idea that bone, itself, exhibits an inflammatory response during diabetes induction. This inflammation could in turn contribute to diabetic bone pathology. IFN‐γ (one of the key cytokines elevated in bone and known to be involved in bone regulation) deficiency did not prevent diabetic bone pathology. Taken together, our findings indicate that bone becomes inflamed with the onset of T1‐diabetes and during this time bone phenotype markers become altered. However, inhibition of one cytokine, IFN‐γ was not sufficient to prevent the rapid bone phenotype changes. J. Cell. Physiol. 218: 575–583, 2009.


Journal of Cellular Physiology | 2004

Hypoxia suppresses runx2 independent of modeled microgravity

Christopher Ontiveros; Regina Irwin; Robert W. Wiseman; Laura R. McCabe

Bone loss is a consequence of skeletal unloading as seen in bed rest and space flight. Unloading decreases oxygenation and osteoblast differentiation/function in bone. Previously we demonstrated that simulation of unloading in vitro, by culturing differentiating mouse osteoblasts in a horizontal rotating wall vessel (RWV), results in suppressed expression of runx2, a master transcriptional regulator of osteoblast differentiation. However, the RWV is able to reproduce in a controlled fashion at least two aspects of disuse that are directly linked, model microgravity and hypoxia. Hypoxia in the RWV is indicated by reduced medium oxygen tension and increased expression of GAPDH and VEGF. To uncouple the role of model microgravity from hypoxia in suppressed runx2 expression, we cultured osteoblasts under modeled microgravity (oxygenated, horizontal RWV rotation), hypoxia (vertical RWV rotation), or both conditions (horizontal RWV rotation). The expression, DNA binding activity and promoter activity of runx2, was suppressed under hypoxic but not normoxic modeled microgravity RWV conditions. Consistent with a role for hypoxia in suppression of runx2, direct exposure to hypoxia alone is sufficient to suppress runx2 expression in osteoblasts grown in standard tissue culture plates. Taken together, our findings indicate that hypoxia associated with skeletal unloading could be major suppressor of runx2 expression leading to suppressed osteoblast differentiation and bone formation.


Inflammatory Bowel Diseases | 2013

Colitis-induced Bone Loss is Gender Dependent and Associated with Increased Inflammation

Regina Irwin; Taehyung Lee; Vincent B. Young; Narayanan Parameswaran; Laura R. McCabe

Background: Patients with inflammatory bowel disease (IBD) are at increase risk for bone loss and fractures. Therefore, in the present study, we examined the effect of experimental IBD on bone health. Methods: We used a murine model of colitis, Helicobacter hepaticus–infected interleukin-10–deficient animals. Molecular and histological properties of bone and intestine were examined to identify the immunopathological consequences of colitis in male and female mice. Results: At 6 weeks postinfection, we observed significant trabecular bone loss in male mice but surprisingly not in female mice. This was true for both distal femur and vertebral locations. In addition, H. hepaticus infection suppressed osteoblast markers only in male mice. Consistent with effects on bone health, male mice with H. hepaticus infection had more severe colitis as determined by histology and elevated levels of inflammatory cytokines in the colon. Although H. hepaticus levels in the stool appeared similar in male and female mice 1 week after infection, by 6 weeks, H. hepaticus levels were greater in male mice, indicating that H. hepaticus survival and virulence within the gastrointestinal tract could be gender dependent. Conclusion: In summary, H. hepaticus–induced colitis severity and associated bone loss is gender regulated, possibly as a result of gender-specific effects on H. hepaticus colonization in the mouse gastrointestinal tract and the consequent immunopathological responses.


Endocrinology | 2015

Loss of Bone and Wnt10b Expression in Male Type 1 Diabetic Mice Is Blocked by the Probiotic Lactobacillus reuteri

Jing Zhang; Katherine J. Motyl; Regina Irwin; Ormond A. MacDougald; Robert A. Britton; Laura R. McCabe

Type 1 diabetes (T1D)-induced osteoporosis is characterized by a predominant suppression of osteoblast number and activity, as well as increased bone marrow adiposity but no change in osteoclast activity. The fundamental mechanisms and alternative anabolic treatments (with few side effects) for T1D bone loss remain undetermined. Recent studies by our laboratory and others indicate that probiotics can benefit bone health. Here, we demonstrate that Lactobacillus reuteri, a probiotic with anti-inflammatory and bone health properties, prevents T1D-induced bone loss and marrow adiposity in mice. We further found that L. reuteri treatment prevented the suppression of Wnt10b in T1D bone. Consistent with a role for attenuated bone Wnt10b expression in T1D osteoporosis, we observed that bone-specific Wnt10b transgenic mice are protected from T1D bone loss. To examine the mechanisms of this protection, we focused on TNF-α, a cytokine up-regulated in T1D that causes suppression of osteoblast Wnt10b expression in vitro. Addition of L. reuteri prevented TNF-α-mediated suppression of Wnt10b and osteoblast maturation markers. Taken together, our findings reveal a mechanism by which T1D causes bone loss and open new avenues for use of probiotics to benefit the bone.


PLOS ONE | 2016

Lactobacillus reuteri 6475 increases bone density in intact females only under an inflammatory setting

Fraser L. Collins; Regina Irwin; Hayley Bierhalter; Jonathan D. Schepper; Robert A. Britton; Narayanan Parameswaran; Laura R. McCabe

Background & Aims We previously demonstrated that short-term oral administration of the probiotic Lactobacillus reuteri 6475 enhanced bone density in male but not female mice. We also established that L. reuteri 6475 enhanced bone health and prevented bone loss in estrogen-deficient female mice. In this study, we tested whether a mild inflammatory state and/or a long-term treatment with the probiotic was required to promote a positive bone effect in estrogen-sufficient female mice. Methods A mild inflammatory state was induced in female mice by dorsal surgical incision (DSI). Following DSI animals were orally supplemented with L. reuteri or vehicle control for a period of 8 weeks. Gene expression was measured in the intestine and bone marrow by qPCR. Distal femoral bone density and architecture was analyzed by micro-CT. Results We report that 8 weeks after DSI there is a significant increase in the weight of spleen, thymus and visceral (retroperitoneal) fat pads. Expression of intestinal cytokines and tight junction proteins are also altered 8 weeks post-DSI. Interestingly, L. reuteri treatment was found to display both intestinal region- and inflammation-dependent effects. Unexpectedly we identified that 1) L. reuteri treatment increased bone density in females but only in those that underwent DSI and 2) DSI benefited cortical bone parameters. In the bone marrow, dorsal surgery induced CD4+ T cell numbers, a response that was unaffected by L. reuteri treatment, whereas expression of RANKL, OPG and IL-10 were significantly affected by L. reuteri treatment. Conclusion Our data reveals a previously unappreciated effect of a mild surgical procedure causing a long-lasting effect on inflammatory gene expression in the gut and the bone. Additionally, we demonstrate that in intact female mice, the beneficial effect of L. reuteri on bone requires an elevated inflammatory status.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2016

Intestinal inflammation without weight loss decreases bone density and growth

Regina Irwin; Sandi Raehtz; Narayanan Parameswaran; Laura R. McCabe

Increasing evidence indicates a strong link between intestinal health and bone health. For example, inflammatory bowel disease can cause systemic inflammation, weight loss, and extra-intestinal manifestations, such as decreased bone growth and density. However, the effects of moderate intestinal inflammation without weight loss on bone health have never been directly examined; yet this condition is relevant not only to IBD but to conditions of increased intestinal permeability and inflammation, as seen with ingestion of high-fat diets, intestinal dysbiosis, irritable bowel syndrome, metabolic syndrome, and food allergies. Here, we induced moderate intestinal inflammation without weight loss in young male mice by treating with a low dose of dextran sodium sulfate (1%) for 15 days. The mice displayed systemic changes marked by significant bone loss and a redistribution of fat from subcutaneous to visceral fat pad stores. Bone loss was caused by reduced osteoblast activity, characterized by decreased expression of osteoblast markers (runx2, osteocalcin), histomorphometry, and dynamic measures of bone formation. In addition, we observed a reduction in growth plate thickness and hypertrophic chondrocyte matrix components (collagen X). Correlation analyses indicate a link between gut inflammation and disease score, but more importantly, we observed that bone density measures negatively correlated with intestinal disease score, as well as colon and bone TNF-α levels. These studies demonstrate that colitis-induced bone loss is not dependent upon weight loss and support a role for inflammation in the link between gut and bone health, an important area for future therapeutic development.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Enhanced production of early lineages of monocytic and granulocytic cells in mice with colitis

Mark D. Trottier; Regina Irwin; Yihang Li; Laura R. McCabe; Pamela J. Fraker

The bone marrow (BM) is a large, highly active, and responsive tissue. Interestingly, little is known about the impact of colitis on hematopoietic functions. Using dextran sodium sulfate (DSS) to induce colitis in mice, we identified significant changes in the BM. Specifically, cells of the monocytic and granulocytic lineages increased nearly 60% and 80%, respectively. This change would support and promote the large infiltration of the gut with neutrophils and monocytes that are the primary cause of inflammation and tissue damage during colitis. Conversely, the early lineages of B and T cells declined in the marrow and thymus with particularly large losses observed among pre-B and pre-T cells with heightened levels of apoptosis noted among CD4+CD8+ thymocytes from DSS-treated mice. Also noteworthy was the 40% decline in cells of the erythrocytic lineages in the marrow of colitis mice, which undoubtedly contributed to the anemia observed in these mice. The peripheral blood reflected the marrow changes as demonstrated by a 2.6-fold increase in neutrophils, a 60% increase in monocytes, and a decline in the lymphocyte population. Thus, colitis changed the BM in profound ways that parallel the general outcomes of colitis including infiltration of the gut with monocytes and neutrophils, inflammation, and anemia. The data provide important understandings of the full impact of colitis that may lead to unique treatments and therapies.


Archive | 2017

Probiotics in gut-bone signaling

Jonathan D. Schepper; Regina Irwin; Jun Kang; Kevin Dagenais; Tristan Lemon; Ally Shinouskis; Narayanan Parameswaran; Laura R. McCabe

The intestinal environment is linked to an array of conditions and diseases, including osteoporosis. Human and animal studies indicate that probiotics can benefit intestinal health and may provide a useful therapeutic to prevent and/or treat bone loss. Probiotics are defined as live microorganisms that when administered in adequate amounts will confer a health benefit on the host. In this review, we will focus on (1) probiotics (definition, history, nomenclature, types), (2) the effects of probiotics on bone health, and (3) mechanisms of probiotic prevention of bone pathologies.

Collaboration


Dive into the Regina Irwin's collaboration.

Top Co-Authors

Avatar

Laura R. McCabe

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert A. Britton

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Darin Quach

Michigan State University

View shared research outputs
Top Co-Authors

Avatar

Jing Zhang

Michigan State University

View shared research outputs
Top Co-Authors

Avatar

Laura Schaefer

Michigan State University

View shared research outputs
Top Co-Authors

Avatar

Taehyung Lee

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge