Network


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

Hotspot


Dive into the research topics where Klara Sjögren is active.

Publication


Featured researches published by Klara Sjögren.


Endocrine Reviews | 2009

The Role of Liver-Derived Insulin-Like Growth Factor-I

Claes Ohlsson; Subburaman Mohan; Klara Sjögren; Åsa Tivesten; Jörgen Isgaard; Olle Isaksson; John-Olov Jansson; Johan Svensson

IGF-I is expressed in virtually every tissue of the body, but with much higher expression in the liver than in any other tissue. Studies using mice with liver-specific IGF-I knockout have demonstrated that liver-derived IGF-I, constituting a major part of circulating IGF-I, is an important endocrine factor involved in a variety of physiological and pathological processes. Detailed studies comparing the impact of liver-derived IGF-I and local bone-derived IGF-I demonstrate that both sources of IGF-I can stimulate longitudinal bone growth. We propose here that liver-derived circulating IGF-I and local bone-derived IGF-I to some extent have overlapping growth-promoting effects and might have the capacity to replace each other (= redundancy) in the maintenance of normal longitudinal bone growth. Importantly, and in contrast to the regulation of longitudinal bone growth, locally derived IGF-I cannot replace (= lack of redundancy) liver-derived IGF-I for the regulation of a large number of other parameters including GH secretion, cortical bone mass, kidney size, prostate size, peripheral vascular resistance, spatial memory, sodium retention, insulin sensitivity, liver size, sexually dimorphic liver functions, and progression of some tumors. It is clear that a major role of liver-derived IGF-I is to regulate GH secretion and that some, but not all, of the phenotypes in the liver-specific IGF-I knockout mice are indirect, mediated via the elevated GH levels. All of the described multiple endocrine effects of liver-derived IGF-I should be considered in the development of possible novel treatment strategies aimed at increasing or reducing endocrine IGF-I activity.


Journal of Bone and Mineral Research | 2012

The gut microbiota regulates bone mass in mice

Klara Sjögren; Cecilia Engdahl; Petra Henning; Ulf H. Lerner; Valentina Tremaroli; Marie K Lagerquist; Fredrik Bäckhed; Claes Ohlsson

The gut microbiota modulates host metabolism and development of immune status. Here we show that the gut microbiota is also a major regulator of bone mass in mice. Germ‐free (GF) mice exhibit increased bone mass associated with reduced number of osteoclasts per bone surface compared with conventionally raised (CONV‐R) mice. Colonization of GF mice with a normal gut microbiota normalizes bone mass. Furthermore, GF mice have decreased frequency of CD4+ T cells and CD11b+/GR 1 osteoclast precursor cells in bone marrow, which could be normalized by colonization. GF mice exhibited reduced expression of inflammatory cytokines in bone and bone marrow compared with CONV‐R mice. In summary, the gut microbiota regulates bone mass in mice, and we provide evidence for a mechanism involving altered immune status in bone and thereby affected osteoclast‐mediated bone resorption. Further studies are required to evaluate the gut microbiota as a novel therapeutic target for osteoporosis.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Estrogen inhibits GH signaling by suppressing GH-induced JAK2 phosphorylation, an effect mediated by SOCS-2

Kin-Chuen Leung; Nathan Doyle; M Ballesteros; Klara Sjögren; Colin K. W. Watts; T H Low; Gary M. Leong; Richard Ross; K. K. Y. Ho

Oral estrogen administration attenuates the metabolic action of growth hormone (GH) in humans. To investigate the mechanism involved, we studied the effects of estrogen on GH signaling through Janus kinase (JAK)2 and the signal transducers and activators of transcription (STATs) in HEK293 cells stably expressing the GH receptor (293GHR), HuH7 (hepatoma) and T-47D (breast cancer) cells. 293GHR cells were transiently transfected with an estrogen receptor-α expression plasmid and luciferase reporters with binding elements for STAT3 and STAT5 or the β-casein promoter. GH stimulated the reporter activities by four- to sixfold. Cotreatment with 17β-estradiol (E2) resulted in a dose-dependent reduction in the response of all three reporters to GH to a maximum of 49–66% of control at 100 nM (P < 0.05). No reduction was seen when E2 was added 1–2 h after GH treatment. Similar inhibitory effects were observed in HuH7 and T-47D cells. E2 suppressed GH-induced JAK2 phosphorylation, an effect attenuated by actinomycin D, suggesting a requirement for gene expression. Next, we investigated the role of the suppressors of cytokine signaling (SOCS) in E2 inhibition. E2 increased the mRNA abundance of SOCS-2 but not SOCS-1 and SOCS-3 in HEK293 cells. The inhibitory effect of E2 was absent in cells lacking SOCS-2 but not in those lacking SOCS-1 and SOCS-3. In conclusion, estrogen inhibits GH signaling, an action mediated by SOCS-2. This paper provides evidence for regulatory interaction between a sex steroid and the GH/JAK/STAT pathway, in which SOCS-2 plays a central mechanistic role.


Nature Medicine | 2014

Osteoblast-derived WNT16 represses osteoclastogenesis and prevents cortical bone fragility fractures.

Sofia Movérare-Skrtic; Petra Henning; Xianwen Liu; Kenichi Nagano; Hiroaki Saito; Anna E. Börjesson; Klara Sjögren; Sara H. Windahl; Helen H. Farman; Bert Kindlund; Cecilia Engdahl; Antti Koskela; Fu-Ping Zhang; Emma Eriksson; Farasat Zaman; Ann Hammarstedt; Hanna Isaksson; Marta Bally; Ali Kassem; Catharina Lindholm; Olof Sandberg; Per Aspenberg; Lars Sävendahl; Jian Q. Feng; Jan Tuckermann; Juha Tuukkanen; Matti Poutanen; Roland Baron; Ulf H. Lerner; Francesca Gori

The WNT16 locus is a major determinant of cortical bone thickness and nonvertebral fracture risk in humans. The disability, mortality and costs caused by osteoporosis-induced nonvertebral fractures are enormous. We demonstrate here that Wnt16-deficient mice develop spontaneous fractures as a result of low cortical thickness and high cortical porosity. In contrast, trabecular bone volume is not altered in these mice. Mechanistic studies revealed that WNT16 is osteoblast derived and inhibits human and mouse osteoclastogenesis both directly by acting on osteoclast progenitors and indirectly by increasing expression of osteoprotegerin (Opg) in osteoblasts. The signaling pathway activated by WNT16 in osteoclast progenitors is noncanonical, whereas the pathway activated in osteoblasts is both canonical and noncanonical. Conditional Wnt16 inactivation revealed that osteoblast-lineage cells are the principal source of WNT16, and its targeted deletion in osteoblasts increases fracture susceptibility. Thus, osteoblast-derived WNT16 is a previously unreported key regulator of osteoclastogenesis and fracture susceptibility. These findings open new avenues for the specific prevention or treatment of nonvertebral fractures, a substantial unmet medical need.


PLOS ONE | 2014

Probiotics protect mice from ovariectomy-induced cortical bone loss.

Claes Ohlsson; Cecilia Engdahl; Frida Fåk; Annica Andersson; Sara H. Windahl; Helen H. Farman; Sofia Movérare-Skrtic; Ulrika Islander; Klara Sjögren

The gut microbiota (GM) modulates the hosts metabolism and immune system. Probiotic bacteria are defined as live microorganisms which when administered in adequate amounts confer a health benefit on the host and can alter the composition of the GM. Germ-free mice have increased bone mass associated with reduced bone resorption indicating that the GM also regulates bone mass. Ovariectomy (ovx) results in bone loss associated with altered immune status. The purpose of this study was to determine if probiotic treatment protects mice from ovx-induced bone loss. Mice were treated with either a single Lactobacillus (L) strain, L. paracasei DSM13434 (L. para) or a mixture of three strains, L. paracasei DSM13434, L. plantarum DSM 15312 and DSM 15313 (L. mix) given in the drinking water during 6 weeks, starting two weeks before ovx. Both the L. para and the L. mix treatment protected mice from ovx-induced cortical bone loss and bone resorption. Cortical bone mineral content was higher in both L. para and L. mix treated ovx mice compared to vehicle (veh) treated ovx mice. Serum levels of the resorption marker C-terminal telopeptides and the urinary fractional excretion of calcium were increased by ovx in the veh treated but not in the L. para or the L. mix treated mice. Probiotic treatment reduced the expression of the two inflammatory cytokines, TNFα and IL-1β, and increased the expression of OPG, a potent inhibitor of osteoclastogenesis, in cortical bone of ovx mice. In addition, ovx decreased the frequency of regulatory T cells in bone marrow of veh treated but not probiotic treated mice. In conclusion, treatment with L. para or the L. mix prevents ovx-induced cortical bone loss. Our findings indicate that these probiotic treatments alter the immune status in bone resulting in attenuated bone resorption in ovx mice.


Journal of Bone and Mineral Research | 2002

Effects of liver-derived insulin-like growth factor I on bone metabolism in mice

Klara Sjögren; Matilda H.-C. Sheng; Sofia Movérare; Jun-Li Liu; Kristina Wallenius; Jan Törnell; Olle Isaksson; John-Olov Jansson; Subburaman Mohan; Claes Ohlsson

Insulin‐like growth factor (IGF) I is an important regulator of both skeletal growth and adult bone metabolism. To better understand the relative importance of systemic IGF‐I versus locally expressed IGF‐I we have developed a transgenic mouse model with inducible specific IGF‐I gene inactivation in the liver (LI‐IGF‐I−/−). These mice are growing normally up to 12 weeks of age but have a disturbed carbohydrate and lipid metabolism. In this study, the long‐term effects of liver‐specific IGF‐I inactivation on skeletal growth and adult bone metabolism were investigated. The adult (week 8–55) axial skeletal growth was decreased by 24% in the LI‐IGF‐I−/− mice whereas no major reduction of the adult appendicular skeletal growth was seen. The cortical cross‐sectional bone area, as measured in the middiaphyseal region of the long bones, was decreased in old LI‐IGF‐I−/− mice. This reduction in the amount of cortical bone was caused mainly by decreased periosteal circumference and was associated with a weaker bone determined by a decrease in ultimate load. In contrast, the amount of trabecular bone was not decreased in the LI‐IGF‐I−/− mice. DNA microarray analysis of 30‐week‐old LI‐IGF‐I−/− and control mice indicated that only four genes were regulated in bone whereas ∼40 genes were regulated in the liver, supporting the hypothesis that liver‐derived IGF‐I is of minor importance for adult bone metabolism. In summary, liver‐derived IGF‐I exerts a small but significant effect on cortical periosteal bone growth and on adult axial skeletal growth while it is not required for the maintenance of the trabecular bone in adult mice.


Endocrinology | 2001

Liver-Derived IGF-I Regulates GH Secretion at the Pituitary Level in Mice

Kristina Wallenius; Klara Sjögren; Xiao Ding Peng; Seungjoon Park; Ville Wallenius; Jun Li Liu; Mia Umaerus; Håkan Wennbo; Olle Isaksson; Lawrence A. Frohman; Rhonda D. Kineman; Claes Ohlsson; John-Olov Jansson

We have reported that liver-specific deletion of IGF-I in mice (LI-IGF-I−/−) results in decreased circulating IGF-I and increased GH levels. In the present study, we determined how elimination of hepatic IGF-I modifies the hypothalamic-pituitary GH axis to enhance GH secretion. The pituitary mRNA levels of GH releasing factor (GHRF) receptor and GH secretagogue (GHS) receptor were increased in LI-IGF-I−/− mice, and in line with this, their GH response to ip injections of GHRF and GHS was increased. Expression of mRNA for pituitary somatostatin receptors, hypothalamic GHRF, somatostatin, and neuropeptide Y was not altered in LI-IGF-I−/− mice, whereas hypothalamic IGF-I expression was increased. Changes in hepatic expression of major urinary protein and the PRL receptor in male LI-IGF-I−/− mice indicated an altered GH release pattern most consistent with enhanced GH trough levels. Liver weight was enhanced in LI-IGF-I−/− mice of both genders. In conclusion, loss of liver-derived IGF-I enhances GH release by...


Proceedings of the National Academy of Sciences of the United States of America | 2013

Estrogen receptor-α in osteocytes is important for trabecular bone formation in male mice

Sara H. Windahl; Anna E. Börjesson; Helen H. Farman; Cecilia Engdahl; Sofia Movérare-Skrtic; Klara Sjögren; Marie K Lagerquist; Jenny M. Kindblom; Antti Koskela; Juha Tuukkanen; Paola Divieti Pajevic; Jian Q. Feng; Karin Dahlman-Wright; Per Antonson; Jan Åke Gustafsson; Claes Ohlsson

The bone-sparing effect of estrogen in both males and females is primarily mediated via estrogen receptor-α (ERα), encoded by the Esr1 gene. ERα in osteoclasts is crucial for the trabecular bone-sparing effect of estrogen in females, but it is dispensable for trabecular bone in male mice and for cortical bone in both genders. We hypothesized that ERα in osteocytes is important for trabecular bone in male mice and for cortical bone in both males and females. Dmp1-Cre mice were crossed with ERαflox/flox mice to generate mice lacking ERα protein expression specifically in osteocytes (Dmp1-ERα−/−). Male Dmp1-ERα−/− mice displayed a substantial reduction in trabecular bone volume (−20%, P < 0.01) compared with controls. Dynamic histomorphometry revealed reduced bone formation rate (−45%, P < 0.01) but the number of osteoclasts per bone surface was unaffected in the male Dmp1-ERα−/− mice. The male Dmp1-ERα−/− mice had reduced expression of several osteoblast/osteocyte markers in bone, including Runx2, Sp7, and Dmp1 (P < 0.05). Gonadal intact Dmp1-ERα−/− female mice had no significant reduction in trabecular bone volume but ovariectomized Dmp1-ERα−/− female mice displayed an attenuated trabecular bone response to supraphysiological E2 treatment. Dmp1-ERα−/− mice of both genders had unaffected cortical bone. In conclusion, ERα in osteocytes regulates trabecular bone formation and thereby trabecular bone volume in male mice but it is dispensable for the trabecular bone in female mice and the cortical bone in both genders. We propose that the physiological trabecular bone-sparing effect of estrogen is mediated via ERα in osteocytes in males, but via ERα in osteoclasts in females.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Roles of transactivating functions 1 and 2 of estrogen receptor-α in bone

Anna E. Börjesson; Sara H. Windahl; Marie K Lagerquist; Cecilia Engdahl; Baruch Frenkel; Sofia Movérare-Skrtic; Klara Sjögren; Jenny M. Kindblom; Alexandra Stubelius; Ulrika Islander; Maria Cristina Antal; Andrée Krust; Pierre Chambon; Claes Ohlsson

The bone-sparing effect of estrogen is primarily mediated via estrogen receptor-α (ERα), which stimulates target gene transcription through two activation functions (AFs), AF-1 in the N-terminal and AF-2 in the ligand binding domain. To evaluate the role of ERα AF-1 and ERα AF-2 for the effects of estrogen in bone in vivo, we analyzed mouse models lacking the entire ERα protein (ERα−/−), ERα AF-1 (ERαAF-10), or ERα AF-2 (ERαAF-20). Estradiol (E2) treatment increased the amount of both trabecular and cortical bone in ovariectomized (OVX) WT mice. Neither the trabecular nor the cortical bone responded to E2 treatment in OVX ERα−/− or OVX ERαAF-20 mice. OVX ERαAF-10 mice displayed a normal E2 response in cortical bone but no E2 response in trabecular bone. Although E2 treatment increased the uterine and liver weights and reduced the thymus weight in OVX WT mice, no effect was seen on these parameters in OVX ERα−/− or OVX ERαAF-20 mice. The effect of E2 in OVX ERαAF-10 mice was tissue-dependent, with no or weak E2 response on thymus and uterine weights but a normal response on liver weight. In conclusion, ERα AF-2 is required for the estrogenic effects on all parameters evaluated, whereas the role of ERα AF-1 is tissue-specific, with a crucial role in trabecular bone and uterus but not cortical bone. Selective ER modulators stimulating ERα with minimal activation of ERα AF-1 could retain beneficial actions in cortical bone, constituting 80% of the skeleton, while minimizing effects on reproductive organs.


Journal of Bone and Mineral Research | 2013

Estrogen receptor-α is required for the osteogenic response to mechanical loading in a ligand-independent manner involving its activation function 1 but not 2.

Sara H. Windahl; Leanne Saxon; Anna E. Börjesson; Marie K Lagerquist; Baruch Frenkel; Petra Henning; Ulf H. Lerner; Gabriel L. Galea; Lee B. Meakin; Cecilia Engdahl; Klara Sjögren; Maria Cristina Antal; Andrée Krust; Pierre Chambon; Lance E. Lanyon; Joanna S. Price; Claes Ohlsson

Estrogen receptor‐α (ERα) is crucial for the adaptive response of bone to loading but the role of endogenous estradiol (E2) for this response is unclear. To determine in vivo the ligand dependency and relative roles of different ERα domains for the osteogenic response to mechanical loading, gene‐targeted mouse models with (1) a complete ERα inactivation (ERα−/−), (2) specific inactivation of activation function 1 (AF‐1) in ERα (ERαAF‐10), or (3) specific inactivation of ERαAF‐2 (ERαAF‐20) were subjected to axial loading of tibia, in the presence or absence (ovariectomy [ovx]) of endogenous E2. Loading increased the cortical bone area in the tibia mainly as a result of an increased periosteal bone formation rate (BFR) and this osteogenic response was similar in gonadal intact and ovx mice, demonstrating that E2 (ligand) is not required for this response. Female ERα−/− mice displayed a severely reduced osteogenic response to loading with changes in cortical area (−78% ± 15%, p < 0.01) and periosteal BFR (−81% ± 9%, p < 0.01) being significantly lower than in wild‐type (WT) mice. ERαAF‐10 mice also displayed a reduced response to mechanical loading compared with WT mice (cortical area −40% ± 11%, p < 0.05 and periosteal BFR −41% ± 8%, p < 0.01), whereas the periosteal osteogenic response to loading was unaffected in ERαAF‐20 mice. Mechanical loading of transgenic estrogen response element (ERE)‐luciferase reporter mice did not increase luciferase expression in cortical bone, suggesting that the loading response does not involve classical genomic ERE‐mediated pathways. In conclusion, ERα is required for the osteogenic response to mechanical loading in a ligand‐independent manner involving AF‐1 but not AF‐2.

Collaboration


Dive into the Klara Sjögren's collaboration.

Top Co-Authors

Avatar

Claes Ohlsson

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar

Olle Isaksson

Sahlgrenska University Hospital

View shared research outputs
Top Co-Authors

Avatar

Sara H. Windahl

Sahlgrenska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Johan Svensson

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge